US8283367B2 - Proteasome inhibitors and methods of using the same - Google Patents

Proteasome inhibitors and methods of using the same Download PDF

Info

Publication number
US8283367B2
US8283367B2 US12/265,998 US26599808A US8283367B2 US 8283367 B2 US8283367 B2 US 8283367B2 US 26599808 A US26599808 A US 26599808A US 8283367 B2 US8283367 B2 US 8283367B2
Authority
US
United States
Prior art keywords
amino
alkyl
methylbutyl
carbonyl
methano
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related, expires
Application number
US12/265,998
Other versions
US20090075936A1 (en
Inventor
Raffaella Bernardini
Alberto Bernareggi
Paolo G. Cassara
Germano D'Arasmo
Ernesto Menta
Ambrogio Oliva
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cephalon LLC
Original Assignee
Cephalon LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cephalon LLC filed Critical Cephalon LLC
Priority to US12/265,998 priority Critical patent/US8283367B2/en
Publication of US20090075936A1 publication Critical patent/US20090075936A1/en
Application granted granted Critical
Publication of US8283367B2 publication Critical patent/US8283367B2/en
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic System
    • C07F5/02Boron compounds
    • C07F5/025Boronic and borinic acid compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to boronic acid and boronic ester compounds useful as proteasome inhibitors and modulation of apoptosis.
  • the proteasome (also referred to as multicatalytic protease (MCP), multicatalytic proteinase, multicatalytic proteinase complex, multicatalytic endopeptidase complex, 20S, 26S, or ingensin) is a large, multiprotein complex present in both the cytoplasm and the nucleus of all eukaryotic cells. It is a highly conserved cellular structure that is responsible for the ATP-dependent proteolysis of most cellular proteins (Tanaka, Biochem Biophy. Res. Commun., 1998, 247, 537).
  • the 26S proteasome consists of a 20S core catalytic complex that is capped at each end by a 19S regulatory subunit.
  • the archaebacterial 20S proteasome contains fourteen copies of two distinct types of subunits, ⁇ and ⁇ , which form a cylindrical structure consisting of four stacked rings.
  • the top and bottom rings contain seven ⁇ -subunits each, while the inner rings contain seven ⁇ -subunits.
  • the more complex eukaryotic 20S proteasome is composed of about 15 distinct 20-30 kDa subunits and is characterized by three major activities with respect to peptide substrates.
  • the proteasome displays tryptic-, chymotryptic-, and peptidylglutamyl peptide-hydrolytic activities (Rivett, Biochem. J., 1993, 291, 1 and Orlowski, Biochemistry, 1990, 29, 10289).
  • the proteasome has a unique active site mechanism which is believed to utilize a threonine residue as the catalytic nucleophile (Seemuller, et al., Science, 1995, 268, 579).
  • the 26S proteasome is able to degrade proteins that have been marked by the addition of ubiquitin molecules.
  • ubiquitin is attached to the 1-amino groups of lysines in a multistep process utilizing ATP and E1 (ubiquitin activating) and E2 (ubiquitin-conjugating) enzymes.
  • Multi-ubiquitinated substrate proteins are recognized by the 26S proteasome and are degraded.
  • the multi-ubiquitin chains are generally released from the complex and ubiquitin is recycled (Goldberg, et al., Nature, 1992, 357, 375).
  • proteasome activity has been implicated in a number of pathologies including neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, as well as occlusion/ischaemia reperfusion injuries, and aging of the central nervous system.
  • the ubiquitin-proteasome pathway also plays a role in neoplastic growth.
  • the regulated degradation of proteins such as cyclins, CDK2 inhibitors, and tumor suppressors is believed to be important in cell cycle progression and mitosis.
  • a known substrate of the proteasome is the tumor suppressor p53 which is involved in several cellular processes (see, e.g., Ko, L. J. Genes Dev., 1996, 10, 1054). Tumor suppressor p53 has been shown to induce apoptosis in several haematopoietic cell lines (Oren, M., Semin. Cancer Biol., 1994, 5, 221).
  • Tumor suppressor p53 degradation is known to be carried out via the ubiquitin-proteasome pathway, and disrupting p53 degradation by inhibition of the proteasome is a possible mode of inducing apoptosis.
  • the proteasome is also required for activation of the transcription factor NF- ⁇ B by degradation of its inhibitory protein, I ⁇ B (Palombella, et al., Cell, 1994, 78, 773).
  • I ⁇ B inhibitory protein
  • NF- ⁇ B has a role in maintaining cell viability through the transcription of inhibitors of apoptosis. Blockade of NF- ⁇ B activity has been demonstrated to make cells more susceptible to apoptosis.
  • Lactacystin is a Streptomyces metabolite that specifically inhibits the proteolytic activity of the proteasome complex (Fenteany, et al., Science, 1995, 268, 726). This molecule is capable of inhibiting the proliferation of several cell types (Fenteany, et al., Proc. Natl. Acad. Sci. USA, 1994, 91, 3358). It has been shown that lactacystin binds irreversibly, through its ⁇ -lactone moiety, to a threonine residue located at the amino terminus of the ⁇ -subunit of the proteasome.
  • Peptide aldehydes have been reported to inhibit the chymotrypsin-like activity associated with the proteasome (Vinitsky, et al., Biochemistry, 1992, 31, 9421; Tsubuki, et al., Biochem. Biophys. Res. Commun., 1993, 196, 1195; and Rock, et al., Cell, 1994, 78, 761).
  • Dipeptidyl aldehyde inhibitors that have IC 50 values in the 10-100 nM range in vitro (Iqbal, M., et al., J. Med. Chem., 1995, 38, 2276) have also been reported.
  • N-terminal peptidyl boronic ester and acid compounds have been reported previously (U.S. Pat. Nos. 4,499,082 and 4,537,773; WO 91/13904; Kettner, et al., J. Biol. Chem., 1984, 259(24), 15106). These compounds are reported to be inhibitors of certain proteolytic enzymes. N-terminal tri-peptide boronic ester and acid compounds have been shown to inhibit the growth of cancer cells (U.S. Pat. No. 5,106,948). A broad class of N-terminal tri-peptide boronic ester and acid compounds and analogs thereof has been shown to inhibit renin (U.S. Pat. No. 5,169,841).
  • Some further proteasome inhibitors can contain boron moieties.
  • Drexler et al., WO 00/64467 report a method of selectively inducing apoptosis in activated endothelial cells or leukemic cells having a high expression level of c-myc by using tetrapeptidic boronate containing proteasome inhibitors.
  • Furet et al., WO 02/096933 report 2-[[N-(2-amino-3-(heteroaryl or aryl)propionyl)aminoacyl]amino]alkylboronic acids and esters for the therapeutic treatment of proliferative diseases in warm-blooded animals.
  • the present invention is directed to novel boronic acid and boronic ester compounds useful as proteasome inhibitors and modulation of apoptosis.
  • the subject invention also comprises methods for inhibition of multicatalytic protease (“MCP”) associated with certain disorders, including the treatment of muscle wasting disorders.
  • MCP multicatalytic protease
  • constituent members are defined infra, as well as preferred constituent members.
  • the present invention provides a compound which is a boronic anhydride of a compound of Formula (I), such as a cyclic boronic anhydride.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula (I) and a pharmaceutically acceptable carrier.
  • the present invention provides a method of inhibiting activity of proteasome comprising contacting a compound of Formula (I) with the proteasome.
  • the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to the cancer a therapeutically effective amount of a compound of Formula (I).
  • the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to the cancer a therapeutically effective amount of a compound of Formula (I), and wherein the cancer is selected from skin, prostate, colorectal, pancreas, kidney, ovary, mammary, liver, tongue, lung, and smooth muscle tissue.
  • the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to the cancer a therapeutically effective amount of a compound of Formula (I), and wherein said cancer is selected from leukemia, lymphoma, non-Hodgkin lymphoma, myeloma, and multiple myeloma.
  • the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to the cancer a therapeutically effective amount of a compound of Formula (I) in combination with one or more antitumor or anticancer agent and/or radiotherapy.
  • the present invention provides a method of inhibiting activity of transcription factor NF- ⁇ B comprising contacting I ⁇ B, the inhibitor of transcription factor NF- ⁇ B, with a compound of Formula (I).
  • the present invention provides a compound of Formula (I) for use in therapy.
  • the present invention provides use of a compound of Formula (I) for the manufacture of a medicament for the treatment of cancer.
  • the present invention provides, inter alia, compounds that can inhibit proteasome activity and be used for the treatment of diseases or disorders related to proteasome activity.
  • Compounds of the invention include compounds of Formula (I):
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH(OH)CH 3 or —CH 2 NR 1a R 1 ;
  • Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R 4 ;
  • R 1 is H, C 1-10 alkyl, carbocyclyl, heterocyclyl, C 1-10 alkyl-C( ⁇ O)—, C 2-10 alkenyl-C( ⁇ O)—, C 2-10 alkynyl-C( ⁇ O)—, carbocyclyl-C( ⁇ O)—, heterocyclyl-C( ⁇ O)—, carbocyclylalkyl-C( ⁇ O)—, heterocyclylalkyl-C( ⁇ O)—, C 1-10 alkyl-S( ⁇ O) 2 —, carbocyclyl-S( ⁇ O) 2 —, heterocyclyl-S( ⁇ O) 2 —, carbocyclylalkyl-S( ⁇ O) 2 —, heterocyclylalkyl-S( ⁇ O) 2 —, C 1 -C 10 alkyl-NHC( ⁇ O)—, carbocyclyl-NHC( ⁇ O)—, heterocyclyl-NHC( ⁇ O)—, carbocyclyl
  • R 1a is H.
  • R 1a and R 1 together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocyclyl group optionally substituted with 1, 2, or 3 R 3 ;
  • R 2 is, independently, H or C 1-6 alkyl
  • two R 2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group;
  • R 3 is, independently, selected from C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —;
  • R 4 is, independently, selected from C 1-20 alkyl, C 2-20 alkenyl, C 2-20 alkynyl, —OR 4a , —SR 4a , —CN, halo, haloalkyl, —NH 2 , —NH(alkyl), —N(alkyl) 2 , —NHC( ⁇ O)O-alkyl, —NHC( ⁇ O)alkyl, —COOH, —C( ⁇ O)O-alkyl, —C( ⁇ O)alkyl, —C(O)H, —S( ⁇ O)-alkyl, —S( ⁇ O) 2 -alkyl, —S( ⁇ O)-aryl, —S( ⁇ O) 2 -aryl, carbocyclyl optionally substituted with 1, 2 or 3 R 5 , and heterocyclyl optionally substituted with 1, 2 or 3 R 5 ;
  • R 4a is H, C 1 -C 20 alkyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, carbocyclyl or heterocyclyl;
  • R 5 is, independently, selected from C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —;
  • Q is boronic acid (B(OH) 2 ) or a cyclic boronic ester wherein said cyclic boronic ester contains from 6 to 10 carbon atoms and contains at least one cycloalkyl moiety.
  • Q is B(OH) 2 or pinanediol boronic ester.
  • Q is pinanediol boronic ester.
  • Z is —CH(OH)CH 3 .
  • Z is —CH 2 NR 1a R 1 .
  • Z is —CH 2 NHR 1 .
  • Z is —CH 2 NHR 1 and R 1 is carbocyclyl-C( ⁇ O)— or carbocyclyl-S( ⁇ O) 2 —, each optionally substituted with 1, 2 or 3 substituents selected from C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, F, Cl, Br, I, C 1-4 haloalkyl, —NH 2 , —NHR 2 , —N(R 2 ) 2 , —N 3 , —NO 2 , —CN, —CNO, —CNS, —C( ⁇ O)OR 2 , —C( ⁇ O)R 2 , —OC( ⁇ O)R 2 , —N(R 2 )C( ⁇ O)R 2 , —N(R 2 )C( ⁇ O)OR 2 , —C( ⁇ O)N(R 2 ) 2 , ureido, —OR 2 , —SR 2 ,
  • Z is —CH 2 NHR 1 and R 1 is aryl-C( ⁇ O)— or aryl-S( ⁇ O) 2 —, each optionally substituted with 1, 2 or 3 substituents selected from C 1-6 alkyl, F, Cl, Br, I, C 1-4 haloalkyl, carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R 3 and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R 3 .
  • Z is —CH 2 NHR 1 and R 1 is phenyl-C( ⁇ O)— or phenyl-S( ⁇ O) 2 —, each optionally substituted with C 1-4 alkyl, F, Cl, Br, I, or aryl.
  • R 1 is aryl-C( ⁇ O)— or aryl-S( ⁇ O) 2 —, each optionally substituted with 1, 2 or 3 substituents selected from C 1-6 alkyl, F, Cl, Br, I, C 1-4 haloalkyl, carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R 3 and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R 3 .
  • R 1 is aryl-C( ⁇ O)— optionally substituted with 1, 2 or 3 substituents selected from C 1-6 alkyl, F, Cl, Br, I, and C 1-4 haloalkyl.
  • R 1 is phenyl-C( ⁇ O)— optionally substituted with 1, 2 or 3 substituents selected from C 1-6 alkyl, F, Cl, Br, I, and C 1-4 haloalkyl.
  • R 1 is —CO-(4-methylphenyl).
  • R 1 is optionally substituted with 1 or 2 substituents.
  • R 1 is optionally substituted with 1 substituent.
  • R 1 is substituted with 1 substituent.
  • R 1 is substituted with C 1-6 alkyl.
  • R 1 is substituted with methyl.
  • Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 R 4 .
  • Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, thiazolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, or indolyl, each optionally substituted by 1, 2 or 3 R 4 .
  • Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, thiazolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, or indolyl, each optionally substituted by 1 or 2 C 1-8 alkyl, carbocyclyl optionally substituted with 1, 2 or 3 R 5 , or heterocyclyl optionally substituted with 1, 2 or 3 R 5 .
  • Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, thiazolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, or indolyl, each optionally substituted by 1 or 2 methyl, ethyl, propyl, butyl, aryl optionally substituted with 1, 2 or 3 R 5 , or heteroaryl optionally substituted with 1, 2 or 3 R 5 .
  • Hy is pyridyl, pyrimidinyl, pyrazinyl, thiazolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, or indolyl, each optionally substituted by 1 or 2 methyl, ethyl, propyl, butyl, aryl optionally substituted with 1, 2 or 3 R 5 , or heteroaryl optionally substituted with 1, 2 or 3 R 5 .
  • Hy is pyrazine substituted by at least 1 or 2 R 4 , unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R 4 , pyridin-4-yl optionally substituted by 1 or 2 R 4 , N-oxo-pyridinyl optionally substituted by 1 or 2 R 4 , pyrimidinyl optionally substituted by 1 or 2 R 4 , imidazolyl optionally substituted by 1 or 2 R 4 , thiazolyl optionally substituted by 1 or 2 R 4 , oxazolyl optionally substituted by 1 or 2 R 4 , pyrrolyl optionally substituted by 1 or 2 R 4 , pyrazolyl optionally substituted by 1 or 2 R 4 , quinolinyl optionally substituted by 1 or 2 R 4 , isoquinolinyl optionally substituted by 1 or 2 R 4 , quinoxalinyl optionally substituted by 1 or 2 R 4 ,
  • Hy is pyrazine substituted by at least 1 or 2 R 4 , unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R 4 , pyridin-4-yl optionally substituted by 1 or 2 R 4 , N-oxo-pyridinyl optionally substituted by 1 or 2 R 4 , pyrimidinyl optionally substituted by 1 or 2 R 4 , imidazolyl optionally substituted by 1 or 2 R 4 , thiazolyl optionally substituted by 1 or 2 R 4 , oxazolyl optionally substituted by 1 or 2 R 4 , pyrrolyl optionally substituted by 1 or 2 R 4 , pyrazolyl optionally substituted by 1 or 2 R 4 , quinolinyl optionally substituted by 1 or 2 R 4 , isoquinolinyl optionally substituted by 1 or 2 R 4 , quinoxalinyl optionally substituted by 1 or 2 R 4 ,
  • Hy is pyrazine substituted by at least 1 or 2 R 4 , unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R 4 , pyridin-4-yl optionally substituted by 1 or 2 R 4 , N-oxo-pyridinyl optionally substituted by 1 or 2 R 4 , pyrimidinyl optionally substituted by 1 or 2 R 4 , imidazolyl optionally substituted by 1 or 2 R 4 , thiazolyl optionally substituted by 1 or 2 R 4 , pyrazolyl optionally substituted by 1 or 2 R 4 , quinolinyl optionally substituted by 1 or 2 R 4 , isoquinolinyl optionally substituted by 1 or 2 R 4 , quinoxalinyl optionally substituted by 1 or 2 R 4 , or indolyl optionally substituted by 1 or 2 R 4 .
  • Hy is pyrazine substituted by at least 1 or 2 R 4 , unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R 4 , pyridin-4-yl optionally substituted by 1 or 2 R 4 , N-oxo-pyridinyl optionally substituted by 1 or 2 R 4 , pyrimidinyl optionally substituted by 1 or 2 R 4 , imidazolyl optionally substituted by 1 or 2 R 4 , thiazolyl optionally substituted by 1 or 2 R 4 , pyrazolyl optionally substituted by 1 or 2 R 4 , quinolinyl optionally substituted by 1 or 2 R 4 , isoquinolinyl optionally substituted by 1 or 2 R 4 , quinoxalinyl optionally substituted by 1 or 2 R 4 , or indolyl optionally substituted by 1 or 2 R 4 , wherein R 4 is C 1-6 alkyl,
  • Hy is pyrazine substituted by at least 1 or 2 R 4 , unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R 4 , pyridin-4-yl optionally substituted by 1 or 2 R 4 , pyrimidinyl optionally substituted by 1 or 2 R 4 , imidazolyl optionally substituted by 1 or 2 R 4 , thiazolyl optionally substituted by 1 or 2 R 4 , pyrazolyl optionally substituted by 1 or 2 R 4 , quinolinyl optionally substituted by 1 or 2 R 4 , isoquinolinyl optionally substituted by 1 or 2 R 4 , quinoxalinyl optionally substituted by 1 or 2 R 4 , or indolyl optionally substituted by 1 or 2 R 4 , wherein R 4 is C 1-6 alkyl, aryl or heterocyclyl.
  • Hy is selected from:
  • R 1a is H.
  • R B is H.
  • R 4 is unsubstituted.
  • R 4 is methyl, ethyl, propyl, butyl, aryl optionally substituted with 1, 2 or 3 R 5 , or heteroaryl optionally substituted with 1, 2 or 3 R 5 .
  • R 4 is C 1-6 alkyl, aryl or heterocyclyl.
  • R 4 is methyl, butyl, phenyl, thienyl, or morpholino.
  • Z is —CH(OH)CH 3 ;
  • Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R 4 .
  • Z is —CH(OH)CH 3 ;
  • Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 R 4 .
  • Z is —CH 2 NHR 1 ;
  • Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R 4 ; and
  • R 1 is carbocyclyl-C( ⁇ O)— or carbocyclyl-S( ⁇ O) 2 —, each optionally substituted with 1, 2 or 3 substituents selected from C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, F, Cl, Br, I, C 1-4 haloalkyl, —NH 2 , —NHR 2 , —N(R 2 ) 2 , —N 3 , —NO 2 , —CN, —CNO, —CNS, —C( ⁇ O)OR 2 , —C( ⁇ O)R 2 , —OC( ⁇ O)R 2 , —N(R 2 )C( ⁇ O)R 2 , —N(R 2 )C( ⁇ O)OR 2 , —C( ⁇ O)N(R 2 ) 2 , ureido, —OR 2 , —SR 2 , —S( ⁇ O)—(C 1-6 alkyl
  • Z is —CH 2 NHR 1 ;
  • Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 R 4 ; and
  • R 1 is carbocyclyl-C( ⁇ O)— or carbocyclyl-S( ⁇ O) 2 —, each optionally substituted with 1, 2 or 3 substituents selected from C 1-6 alkyl, F, Cl, Br, I, C 1-4 haloalkyl, carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R 3 , and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R 3 .
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH(OH)CH 3 or —CH 2 NHR 1 ;
  • Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 R 4 ;
  • R 1 is carbocyclyl-C( ⁇ O)— or heterocyclyl-C( ⁇ O)—, each optionally substituted with 1, 2 or 3 R 3 ;
  • R 3 is, independently, selected from C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —;
  • R 4 is, independently, selected from C 1-6 alkyl, aryl and heterocyclyl
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH(OH)CH 3 or —CH 2 NHR 1 ;
  • Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 C 1-6 alkyl, aryl or heterocyclyl;
  • R 1 is carbocyclyl-C( ⁇ O)— or heterocyclyl-C( ⁇ O)—, each optionally substituted with 1, 2 or 3 R 3 ;
  • R 3 is, independently, selected from C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —;
  • R 4 is, independently, selected from C 1-6 alkyl, aryl and heterocyclyl
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH(OH)CH 3 or —CH 2 NHR 1 ;
  • Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 C1-6 alkyl, phenyl, thienyl or morpholino;
  • R 1 is carbocyclyl-C( ⁇ O)— or heterocyclyl-C( ⁇ O)—, each optionally substituted with 1, 2 or 3 R 3 ;
  • R 3 is, independently, selected from C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —;
  • R 4 is, independently, selected from C 1-6 alkyl, aryl and heterocyclyl
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH(OH)CH 3 or —CH 2 NHR 1 ;
  • Hy is selected from
  • R 1 is carbocyclyl-C( ⁇ O)— or heterocyclyl-C( ⁇ O)—, each optionally substituted with 1, 2 or 3 R 3 ;
  • R 3 is, independently, selected from C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —.
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH(OH)CH 3 or —CH 2 NHR 1 ;
  • Hy is selected from
  • R 1 is —CO-(4-methylphenyl).
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OH) 2 or pinanediol boronic ester
  • Z is —CH(OH)CH 3 or —CH 2 NHR 1 ;
  • Hy is selected from
  • R 1 is —CO-(4-methylphenyl).
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH(OH)CH 3 ;
  • Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R 4 ;
  • R B is, independently, selected from C 1-20 alkyl, C 2-20 alkenyl, C 2-20 alkynyl, —OR 4a , SR 4a , —CN, halo, haloalkyl, —NH 2 , —NH(alkyl), —N(alkyl) 2 , —NHC( ⁇ O)O-alkyl, —NHC( ⁇ O)alkyl, —COOH, —C( ⁇ O)O-alkyl, —C( ⁇ O)alkyl, —C(O)H, —S( ⁇ O)-alkyl, —S( ⁇ O) 2 -alkyl, —S( ⁇ O)-aryl, —S( ⁇ O) 2 -aryl, carbocyclyl optionally substituted with 1, 2 or 3 R 5 , and heterocyclyl optionally substituted with 1, 2 or 3 R 5 ;
  • R 4a is H, C 1 -C 20 alkyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, carbocyclyl or heterocyclyl;
  • R 5 is, independently, selected from C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —;
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH(OH)CH 3 ;
  • Hy is pyrazine substituted by at least 1 or 2 R 4 , unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R 4 , pyridin-4-yl optionally substituted by 1 or 2 R 4 , N-oxo-pyridinyl optionally substituted by 1 or 2 R 4 , pyrimidinyl optionally substituted by 1 or 2 R 4 , imidazolyl optionally substituted by 1 or 2 R 4 , thiazolyl optionally substituted by 1 or 2 R 4 , oxazolyl optionally substituted by 1 or 2 R 4 , pyrrolyl optionally substituted by 1 or 2 R 4 , pyrazolyl optionally substituted by 1 or 2 R 4 , quinolinyl optionally substituted by 1 or 2 R 4 , isoquinolinyl optionally substituted by 1 or 2 R 4 , quinoxalinyl optionally substituted by 1 or 2 R 4 ,
  • R 4 is, independently, selected from C 1-20 alkyl, C 2-20 alkenyl, C 2-20 alkynyl, —OR 4a , —SR 4a , —CN, halo, haloalkyl, —NH 2 , —NH(alkyl), —N(alkyl) 2 , —NHC( ⁇ O)O-alkyl, —NHC( ⁇ O)alkyl, —COOH, —C( ⁇ O)O-alkyl, —C( ⁇ O)alkyl, —C(O)H, —S( ⁇ O)-alkyl, —S( ⁇ O) 2 -alkyl, —S( ⁇ O)-aryl, —S( ⁇ O) 2 -aryl, carbocyclyl optionally substituted with 1, 2 or 3 R 5 , and heterocyclyl optionally substituted with 1, 2 or 3 R 5 ;
  • R 4a is H, C 1 -C 20 alkyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, carbocyclyl or heterocyclyl;
  • R 5 is, independently, selected from C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —.
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH(OH)CH 3 ;
  • Hy is pyrazine substituted by at least 1 or 2 R 4 , unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R 4 , pyridin-4-yl optionally substituted by 1 or 2 R 4 , N-oxo-pyridinyl optionally substituted by 1 or 2 R 4 , pyrimidinyl optionally substituted by 1 or 2 R 4 , imidazolyl optionally substituted by 1 or 2 R 4 , thiazolyl optionally substituted by 1 or 2 R 4 , pyrazolyl optionally substituted by 1 or 2 R 4 , quinolinyl optionally substituted by 1 or 2 R 4 , isoquinolinyl optionally substituted by 1 or 2 R 4 , quinoxalinyl optionally substituted by 1 or 2 R 4 , or indolyl optionally substituted by 1 or 2 R 4 ;
  • R 4 is, independently, selected from C 1-20 alkyl, C 2-20 alkenyl, C 2-20 alkynyl, —OR 4a , —SR 4a , —CN, halo, haloalkyl, —NH 2 , —NH(alkyl), —N(alkyl) 2 , —NHC( ⁇ O)O-alkyl, —NHC( ⁇ O)alkyl, —COOH, —C( ⁇ O)O-alkyl, —C( ⁇ O)alkyl, —C(O)H, —S( ⁇ O)-alkyl, —S( ⁇ O) 2 -alkyl, —S( ⁇ O)-aryl, —S( ⁇ O) 2 -aryl, carbocyclyl optionally substituted with 1, 2 or 3 R 5 , and heterocyclyl optionally substituted with 1, 2 or 3 R 5 ;
  • R 4a is H, C 1 -C 20 alkyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, carbocyclyl or heterocyclyl;
  • R 5 is, independently, selected from C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —.
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH(OH)CH 3 ;
  • Hy is selected from
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OH) 2 or pinanediol boronic ester
  • Z is —CH(OH)CH 3 ;
  • Hy is selected from
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH 2 NR 1a R 1 ;
  • Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R 4 ;
  • R 1 is H, C 1-10 alkyl, carbocyclyl, heterocyclyl, C 1-10 alkyl-C( ⁇ O)—, C 2-10 alkenyl-C( ⁇ O)—, C 2-10 alkynyl-C( ⁇ O)—, carbocyclyl-C( ⁇ O)—, heterocyclyl-C( ⁇ O)—, carbocyclylalkyl-C( ⁇ O)—, heterocyclylalkyl-C( ⁇ O)—, C 1-10 alkyl-S( ⁇ O) 2 —, carbocyclyl-S( ⁇ O) 2 —, heterocyclyl-S( ⁇ O) 2 —, carbocyclylalkyl-S( ⁇ O) 2 —, heterocyclylalkyl-S( ⁇ O) 2 —, C 1 -C 10 alkyl-NHC( ⁇ O)—, carbocyclyl-NHC( ⁇ O)—, heterocyclyl-NHC( ⁇ O)—, carbocyclyl
  • R 1a is H.
  • R 1a and R 1 together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocyclyl group optionally substituted with 1, 2, or 3 R 3 ;
  • R 2 is, independently, H or C 1-6 alkyl
  • two R 2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group;
  • R 3 is, independently, selected from C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —;
  • R 4 is, independently, selected from C 1-20 alkyl, C 2-20 alkenyl, C 2-20 alkynyl, —OR 4a , —SR 4a , —CN, halo, haloalkyl, —NH 2 , —NH(alkyl), —N(alkyl) 2 , —NHC( ⁇ O)O-alkyl, —NHC( ⁇ O)alkyl, —COOH, —C( ⁇ O)O-alkyl, —C( ⁇ O)alkyl, —C(O)H, —S( ⁇ O)-alkyl, —S( ⁇ O) 2 -alkyl, —S( ⁇ O)-aryl, —S( ⁇ O) 2 -aryl, carbocyclyl optionally substituted with 1, 2 or 3 R 5 , and heterocyclyl optionally substituted with 1, 2 or 3 R 5 ;
  • R 4a is H, C 1 -C 20 alkyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, carbocyclyl or heterocyclyl;
  • R 5 is, independently, selected from C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —.
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH 2 NR 1a R 1 ;
  • Hy is pyrazine substituted by at least 1 or 2 R 4 , pyridinyl optionally substituted by 1 or 2 R 4 , N-oxo-pyridinyl optionally substituted by 1 or 2 R 4 , pyrimidinyl optionally substituted by 1 or 2 R 4 , imidazolyl optionally substituted by 1 or 2 R 4 , thiazolyl optionally substituted by 1 or 2 R 4 , oxazolyl optionally substituted by 1 or 2 R 4 , pyrrolyl optionally substituted by 1 or 2 R 4 , pyrazolyl optionally substituted by 1 or 2 R 4 , quinolinyl optionally substituted by 1 or 2 R 4 , isoquinolinyl optionally substituted by 1 or 2 R 4 , quinoxalinyl optionally substituted by 1 or 2 R 4 , indolyl optionally substituted by 1 or 2 R 4 , quinazolinyl optionally substituted by 1 or 2 R 4 ,
  • R 1 is H, C 1-10 alkyl, carbocyclyl, heterocyclyl, C 1-10 alkyl-C( ⁇ O)—, C 2-10 alkenyl-C( ⁇ O)—, C 2-10 alkynyl-C( ⁇ O)—, carbocyclyl-C( ⁇ O)—, heterocyclyl-C( ⁇ O)—, carbocyclylalkyl-C( ⁇ O)—, heterocyclylalkyl-C( ⁇ O)—, C 1-10 alkyl-S( ⁇ O) 2 —, carbocyclyl-S( ⁇ O) 2 —, heterocyclyl-S( ⁇ O) 2 —, carbocyclylalkyl-S( ⁇ O) 2 —, heterocyclylalkyl-S( ⁇ O) 2 —, C 1 -C 10 alkyl-NHC( ⁇ O)—, carbocyclyl-NHC( ⁇ O)—, heterocyclyl-NHC( ⁇ O)—, carbocyclyl
  • R 1a is H.
  • R 1a and R 1 together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocyclyl group optionally substituted with 1, 2, or 3 R 3 ;
  • R 2 is, independently, H or C 1-6 alkyl
  • two R 2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group;
  • R 3 is, independently, selected from C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —;
  • R 4 is, independently, selected from C 1-20 alkyl, C 2-20 alkenyl, C 2-20 alkynyl, —OR 4a , SR 4a , —CN, halo, haloalkyl, —NH 2 , —NH(alkyl), —N(alkyl) 2 , —NHC( ⁇ O)O-alkyl, —NHC( ⁇ O)alkyl, —COOH, —C( ⁇ O)O-alkyl, —C( ⁇ O)alkyl, —C(O)H, —S( ⁇ O)-alkyl, —S( ⁇ O) 2 -alkyl, —S( ⁇ O)-aryl, —S( ⁇ O) 2 -aryl, carbocyclyl optionally substituted with 1, 2 or 3 R 5 , and heterocyclyl optionally substituted with 1, 2 or 3 R 5 ;
  • R 4a is H, C 1 -C 20 alkyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, carbocyclyl or heterocyclyl;
  • R 5 is, independently, selected from C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —.
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH 2 NR 1a R 1 ;
  • Hy is selected from
  • R 1 is H, C 1-10 alkyl, carbocyclyl, heterocyclyl, C 1-10 alkyl-C( ⁇ O)—, C 2-10 alkenyl-C( ⁇ O)—, C 2-10 alkynyl-C( ⁇ O)—, carbocyclyl-C( ⁇ O)—, heterocyclyl-C( ⁇ O)—, carbocyclylalkyl-C( ⁇ O)—, heterocyclylalkyl-C( ⁇ O)—, C 1-10 alkyl-S( ⁇ O) 2 —, carbocyclyl-S( ⁇ O) 2 —, heterocyclyl-S( ⁇ O) 2 —, carbocyclylalkyl-S( ⁇ O) 2 —, heterocyclylalkyl-S( ⁇ O) 2 —, C 1 -C 10 alkyl-NHC( ⁇ O)—, carbocyclyl-NHC( ⁇ O)—, heterocyclyl-NHC( ⁇ O)—, carbocyclyl
  • R 1a is H.
  • R 1a and R 1 together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocyclyl group optionally substituted with 1, 2, or 3 R 3 ;
  • R 2 is, independently, H or C 1-6 alkyl
  • two R 2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group
  • R 3 is, independently, selected from C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —.
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH 2 NHR 1 ;
  • Hy is selected from
  • R 1 is H, C 1-10 alkyl, carbocyclyl, heterocyclyl, C 1-10 alkyl-C( ⁇ O)—, C 2-10 alkenyl-C( ⁇ O)—, C 2-10 alkynyl-C( ⁇ O)—, carbocyclyl-C( ⁇ O)—, heterocyclyl-C( ⁇ O)—, carbocyclylalkyl-C( ⁇ O)—, heterocyclylalkyl-C( ⁇ O)—, C 1-10 alkyl-S( ⁇ O) 2 —, carbocyclyl-S( ⁇ O) 2 —, heterocyclyl-S( ⁇ O) 2 —, carbocyclylalkyl-S( ⁇ O) 2 —, heterocyclylalkyl-S( ⁇ O) 2 —, C 1 -C 10 alkyl-NHC( ⁇ O)—, carbocyclyl-NHC( ⁇ O)—, heterocyclyl-NHC( ⁇ O)—, carbocyclyl
  • R 2 is, independently, H or C 1-6 alkyl
  • two R 2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group
  • R 3 is, independently, selected from C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —.
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH 2 NHR 1 ;
  • Hy is selected from
  • R 1 is carbocyclyl-C( ⁇ O)—, heterocyclyl-C( ⁇ O)—, wherein R 1 is optionally substituted with 1, 2 or 3 substituents selected from C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, F, Cl, Br, I, C 1-4 haloalkyl, —NH 2 , —NHR 2 , —N(R 2 ) 2 , —N 3 , —NO 2 , —CN, —CNO, —CNS, —C( ⁇ O)OR 2 , —C( ⁇ O)R 2 , —OC( ⁇ O)R 2 , —N(R 2 )C( ⁇ O)R 2 , —N(R 2 )C( ⁇ O)OR 2 , —C( ⁇ O)N(R 2 ) 2 , ureido, —OR 2 , —SR 2 , —S( ⁇ O)—(C 1-6 alky
  • R 2 is, independently, H or C 1-6 alkyl
  • two R 2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group
  • R 3 is, independently, selected from C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and H 2 NS( ⁇ O) 2 —.
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH 2 NHR 1 ;
  • Hy is selected from
  • R 1 is aryl-C( ⁇ O)—, wherein R 1 is optionally substituted with 1 or 2 substituents selected from C 1-6 alkyl, F, Cl, Br, I, and C 1-4 haloalkyl.
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OR B ) 2 , boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH 2 NHR 1 ;
  • Hy is selected from
  • R 1 is —CO-(4-methylphenyl).
  • compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(OH) 2 pinanediol boronic ester
  • Z is —CH 2 NHR 1 ;
  • Hy is selected from
  • R 1 is —CO-(4-methylphenyl).
  • compounds of the invention include compounds of Formula (I):
  • R B is, independently, H, C 1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
  • Z is —CH(OH)CH 3 or —CH 2 NR 1a R 1 ;
  • Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R 4 ;
  • R 1 is H, C 1-10 alkyl, carbocyclyl, heterocyclyl, C 1-10 alkyl-C( ⁇ O)—, C 2-10 alkenyl-C( ⁇ O)—, C 2-10 alkynyl-C( ⁇ O)—, carbocyclyl-C( ⁇ O)—, heterocyclyl-C( ⁇ O)—, carbocyclylalkyl-C( ⁇ O)—, heterocyclylalkyl-C( ⁇ O)—, C 1-10 alkyl-S( ⁇ O) 2 —, carbocyclyl-S( ⁇ O) 2 —, heterocyclyl-S( ⁇ O) 2 —, carbocyclylalkyl-S( ⁇ O) 2 —, heterocyclylalkyl-S( ⁇ O) 2 —, C 1 -C 10 alkyl-NHC( ⁇ O)—, carbocyclyl-NHC( ⁇ O)—, heterocyclyl-NHC( ⁇ O)—, carbocyclyl
  • R 1a is H.
  • R 1a and R 1 together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocyclyl group optionally substituted with 1, 2, or 3 R 3 ;
  • R 2 is, independently, H or C 1-6 alkyl
  • two R 2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group;
  • R 3 is, independently, selected from C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, (alkyl-O) r -alkyl, HO-(alkyl-O) r -alkyl-, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and
  • R 4 is, independently, selected from C 1-20 alkyl, C 2-20 alkenyl, C 2-20 alkynyl, —OR 4a , —SR 4a , —CN, halo, haloalkyl, —NH 2 , —NH(alkyl), —N(alkyl) 2 , —NHC( ⁇ O)O-alkyl, —NHC( ⁇ O)alkyl, —COOH, —C( ⁇ O)O-alkyl, —C( ⁇ O)alkyl, —C(O)H, —S( ⁇ O)-alkyl, —S( ⁇ O) 2 -alkyl, —S( ⁇ O)-aryl, —S( ⁇ O) 2 -aryl, carbocyclyl optionally substituted with 1, 2 or 3 R 5 , and heterocyclyl optionally substituted with 1, 2 or 3 R 5 ;
  • R 4a is H, C 1 -C 20 alkyl, C 2 -C 20 alkenyl, C 2 -C 20 alkynyl, carbocyclyl or heterocyclyl;
  • R 5 is, independently, selected from C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, alkyl-OC( ⁇ O)NH—, aryl-OC( ⁇ O)NH—, alkyl-C( ⁇ O)NH—, alkyl-C( ⁇ O)O—, (alkyl-O) r -alkyl, HO-(alkyl-O) r -alkyl-, —OH, —SH, —CN, —N 3 , —CNO, —CNS, alkyl-S( ⁇ O)—, alkyl-S( ⁇ O) 2 —, H 2 NS( ⁇ O)—, and
  • r 0, 1, 2, 3 or 4;
  • boronic acid refers to a compound containing a B(OH) 2 moiety.
  • boronic acid compounds can form oligomeric anhydrides by dehydration of the boronic moiety.
  • Snyder, et al., J. Am. Chem. Soc., 1958, 80, 3611 report oligomeric arylboronic acids.
  • “boronic acid”, or a chemical formula containing a —B(OH) 2 moiety is intended to encompass free boronic acids, oligomeric anhydrides, including but not limited to, dimers, trimers, tetramers, and mixtures thereof.
  • boronic acid anhydride or “boronic anhydride” refers to a compound formed by the combination of two or more molecules of a boronic acid compound of Formula (I), with loss of one or more water molecules from the boronic acid moieties. When contacted with water, the boronic acid anhydride compound can be hydrated to release free boronic acid compound.
  • the boronic acid anhydride structure can contain two, three, four, or more boronic acid units and can have a cyclic or linear configuration.
  • the boronic acid anhydride compound exists substantially in a single oligomeric form; however, boronic acid anhydrides also encompass mixtures of different oligomeric boronic acid anhydride as well as free boronic acids.
  • Non-limiting examples of boronic acid anhydrides of the invention include compounds of Formula (II) and (III) where G is a moiety of Formula (IV) and t is 0 to 10 or 1, 2, 3, or 4.
  • At least about 80% of boronic acid present in a boronic acid anhydride compound exists in a single oligomeric anhydride form. In further embodiments, at least about 85, about 90, about 95, or about 99% of the boronic acid present in the boronic acid anhydride exists in a single oligomeric anhydride form.
  • the boronic acid anhydride compound consists essentially of a single oligomeric boronic acid anhydride.
  • the boronic acid anhydride compound consists of a single oligomeric boronic acid anhydride.
  • the boronic acid anhydride compound contains a boroxine of Formula (III), wherein t is 1.
  • Boronic acid anhydride compounds can be prepared from the corresponding boronic acid compound by exposure to dehydrating conditions, including, for example, crystallization, lyophilization, exposure to heat, and/or exposure to a drying agent.
  • suitable crystallization solvents include ethyl acetate, dichloromethane, hexanes, ether, benzene, acetonitrile, ethanol, and mixtures thereof.
  • boronic ester or “boronic acid ester” refers to an ester derivative of a boronic acid compound.
  • cyclic boronic ester is intended to mean a stable cyclic boronic moiety of general formula —B(OR)(OR) wherein the two R substituents are linked together forming a cyclic moiety (e.g., 3- to 10-membered cycloalkyl group) optionally further substituted with one or more substituents or fused with (sharing at least one bond) one or more further carbocyclyl or heterocarbocyclyl groups.
  • the cyclic boronic ester can contain from 2 to 20 carbon atoms, and optionally, a heteroatom which can be N, S, or O.
  • Cyclic boronic esters are well known in the art. Examples of cyclic boronic esters include, but are not limited to, pinanediol boronic ester, pinacol boronic ester, 1,2-ethanediol boronic ester, 1,3-propanediol boronic ester, 1,2-propanediol boronic ester, 2,3-butanediol boronic ester, 1,1,2,2-tetramethylethanediol boronic ester, 1,2-diisopropylethanediol boronic ester, 5,6-decanediol boronic ester, 1,2-dicyclohexylethanediol boronic ester, bicyclohexyl-1,1′-diol, di
  • the “cyclic boronic ester” has Formula (II-a):
  • D is absent, O, S, NR 16 , or CR 15e R 15f ,
  • R 15a , R 15b , R 15c , R 15d , R 15e , R 15f are each, independently, H, C 1 -C 10 alkyl C 3 -C 7 cycloalkyl, aryl or heteroaryl, wherein said C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, aryl or heteroaryl are each optionally substituted by 1, 2, 3 or 4 halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, C 1 -C 4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl;
  • R 15a and R 15b together with the C atoms to which they are attached form C 3 -C 10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, C 1 -C 4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl;
  • R 15c and R 15d together with the C atoms to which they are attached form C 3 -C 10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, C 1 -C 4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl;
  • R 15b and R 15c together with the C atoms to which they are attached and the intevening D moiety form aryl, heteroaryl, C 3 -C 10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, C 1 -C 4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl;
  • R 16 is H or C 1 -C 6 alkyl
  • p and q are each, independently, 1, 2 or 3.
  • D is absent.
  • D is NR 16 .
  • D is NH
  • D is CH 2 .
  • R 15a and R 15b together with the C atoms to which they are attached form C 3 -C 10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, C 1 -C 4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; and R 15c and R 15d together with the C atoms to which they are attached form C 3 -C 10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, C 1 -C 4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl.
  • R 15a and R 15b together with the C atoms to which they are attached form cyclopropyl, cyclobutyl, cyclopenytyl, cyclohexyl or cycloheptyl; and R 15c and R 15d together with the C atoms to which they are attached form cyclopropyl, cyclobutyl, cyclopenytyl, cyclohexyl or cycloheptyl.
  • D is absent and R 15b and R 15c together with the C atoms to which they are attached form aryl, heteroaryl, C 3 -C 10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, C 1 -C 4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl.
  • D is absent and R 15b and R 15c together with the C atoms to which they are attached form C 3 -C 10 cycloalkyl optionally substituted by 1, 2, 3 or 4 halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, C 1 -C 4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl.
  • D is absent and R 15b and R 15c together with the C atoms to which they are attached form C 3 -C 10 cycloalkyl optionally substituted by 1, 2, 3 or 4 halo or C 1 -C 4 alkyl.
  • D is absent and R 15b and R 15c together with the C atoms to which they are attached form a C 7 -C 10 bicyclic cycloalkyl group optionally substituted by 1, 2, 3 or 4 halo or C 1 -C 4 alkyl.
  • p and q are each 1.
  • At least one of R 15a , R 15b , R 15c , R 15d is other than H.
  • cyclic boronic esters include, boronic esters with the following structures:
  • W is a substituted or unsubstituted C 4 -C 10 cycloalkyl ring or a substituted or unsubstituted phenyl ring; W 1 is, independently at each occurrence, a substituted or unsubstituted C 3 -C 6 cycloalkyl ring.
  • Groups R 15a , R 15b , R 15c , R 15d , R 15e , R 15f , p and q are, defined as provided above.
  • alkyl or “alkylene” is meant to refer to a saturated hydrocarbon group which is straight-chained or branched.
  • Example alkyl groups include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, s-butyl, t-butyl), pentyl (e.g., n-pentyl, isopentyl, neopentyl) and the like.
  • An alkyl group can contain from 1 to about 20, from 2 to about 20, from 1 to about 10, from 1 to about 8, from 1 to about 6, from 1 to about 4, or from 1 to about 3 carbon atoms.
  • alkenyl refers to an alkyl group having one or more double carbon-carbon bonds.
  • Example alkenyl groups include ethenyl, propenyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl, and the like.
  • alkynyl refers to an alkyl group having one or more triple carbon-carbon bonds.
  • Example alkynyl groups include ethynyl, propynyl, butynyl, pentynyl, and the like.
  • haloalkyl refers to an alkyl group having one or more halogen substituents.
  • Example haloalkyl groups include CF 3 , C 2 F 5 , CHF 2 , CCl 3 , CHCl 2 , C 2 Cl 5 , and the like.
  • An alkyl group in which all of the hydrogen atoms are replaced with halogen atoms can be referred to as “perhaloalkyl.”
  • perhaloalkyl groups include CF 3 and C 2 F 5 .
  • Carbocyclyl groups are saturated (i.e., containing no double or triple bonds) or unsaturated (i.e., containing one or more double or triple bonds) cyclic hydrocarbon moieties. Carbocyclyl groups can be mono- or polycyclic.
  • Example carbocyclyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, 1,3-cyclopentadienyl, cyclohexenyl, norbornyl, norpinyl, norcarnyl, adamantyl, phenyl, and the like.
  • Carbocyclyl groups can be aromatic (e.g., “aryl”) or non-aromatic (e.g., “cycloalkyl”). In some embodiments, carbocyclyl groups can have from 3 to about 20, 3 to about 10, or 3 to about 7 carbon atoms.
  • aryl refers to aromatic carbocyclyl groups including monocyclic or polycyclic aromatic hydrocarbons such as, for example, phenyl, naphthyl, anthracenyl, phenanthrenyl, indanyl, indenyl, and the like. In some embodiments, aryl groups have from 6 to about 18 ring-forming carbon atoms.
  • cycloalkyl refers to non-aromatic carbocyclyl groups including cyclized alkyl, alkenyl, and alkynyl groups. Cycloalkyl groups can include bi- or poly-cyclic ring systems. Example cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptatrienyl, norbornyl, norpinyl, norcarnyl, adamantyl, and the like.
  • cycloalkyl moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the cycloalkyl ring, for example, benzo derivatives of cyclopentane (indanyl), cyclohexane (tetrahydronaphthyl), and the like.
  • groups in which one or more of the ring-forming carbon atoms is substituted by an oxo group can have 3, 4, 5, 6, or 7 ring forming carbon atoms.
  • cycloalkyl groups can have 0, 1, or 2 double or triple ring-forming bonds.
  • heterocyclyl groups can be saturated or unsaturated carbocyclyl groups wherein one or more of the ring-forming carbon atoms of the carbocyclyl group is replaced with a heteroatom such as O, S, or N.
  • Heterocyclyl groups can be aromatic (e.g., “heteroaryl”) or non-aromatic (e.g., “heterocycloalkyl”).
  • Heterocyclyl groups can correspond to hydrogenated and partially hydrogenated heteroaryl groups.
  • Heterocyclyl groups can contain, in addition to at least one heteroatom, from about 1 to about 20, about 2 to about 10, or about 2 to about 7 carbon atoms and can be attached through a carbon atom or heteroatom.
  • any ring-forming carbon atom or heteroatom can be substituted by one or two oxo or sulfide groups.
  • heterocyclyl groups include morpholino, thiomorpholino, piperazinyl, tetrahydrofuranyl, tetrahydrothienyl, 2,3-dihydrobenzofuryl, 1,3-benzodioxole, benzo-1,4-dioxane, piperidinyl, pyrrolidinyl, isoxazolidinyl, isothiazolidinyl, pyrazolidinyl, oxazolidinyl, thiazolidinyl, imidazolidinyl, and the like.
  • the heterocyclyl group is a 5- or 6-membered heterocyclyl group.
  • heteroaryl groups are aromatic heterocarbocyclyl groups and include monocyclic and polycyclic aromatic hydrocarbons that have at least one heteroatom ring member such as sulfur, oxygen, or nitrogen.
  • Heteroaryl groups include, without limitation, pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrrolyl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl indazolyl, 1,2,4-thiadiazolyl, isothiazolyl, benzothienyl, purinyl, carbazolyl, benzimidazolyl, pyrimidin
  • heteroaryl groups can have from 3 to about 20 ring-forming carbon atoms, and in further embodiments from about 3 to about 12 ring forming carbon atoms. In some embodiments, heteroaryl groups have 1 to about 4, 1 to about 3, or 1 to 2 heteroatoms. In some embodiments, the heteroaryl group has at least one ring-forming N atom.
  • heterocycloalkyl refers to a non-aromatic heterocyclyl group including cyclized alkyl, alkenyl, and alkynyl groups where one or more of the ring-forming carbon atoms is replaced by a heteroatom such as an O, N, or S atom.
  • Ring-forming carbon and heteroatoms such as S and N can further be oxidized in a heterocycloalkyl moeity.
  • the ring-forming carbon or heteroatom can bear one or two oxo or sufido moieties (e.g., >C ⁇ O, >S ⁇ O, >S( ⁇ O) 2 , N ⁇ O, etc.).
  • heterocycloalkyl moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the nonaromatic heterocyclic ring, for example phthalimidyl, naphthalimidyl pyromellitic diimidyl, phthalanyl, and benzo derivatives of saturated heterocycles such as indolene and isoindolene groups.
  • heterocycloalkyl groups have 3 to about 20 ring-forming atoms.
  • heterocycloalkyl groups have 3, 4, 5, 6, or 7 ring-forming atoms.
  • heterocycloalkyl groups have 0, 1, or 2 double or triple ring-forming bonds.
  • halo or “halogen” includes fluoro, chloro, bromo, and iodo.
  • alkoxy refers to an —O-alkyl group.
  • Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the like. In some embodiments, alkoxy groups have from 1 to 20, 1 to 12, 1 to 8, 1 to 6, 1 to 4 or 1 to 3 carbon atoms.
  • alkoxyalkoxy refers to an —O-alkyl-O-alkyl group.
  • thioalkoxy refers to an alkoxy group in which the O atom is replaced by an S atom.
  • aryloxy refers to an —O-aryl group.
  • An example aryloxy group is phenoxy.
  • thioaryloxy refers to an aryloxy group in which the O atom is replaced by an S atom.
  • aralkyl refers to an alkyl moiety substituted by an aryl group.
  • Example aralkyl groups include benzyl and naphthylmethyl groups. In some embodiments, aralkyl groups have from 7 to 11 carbon atoms.
  • amino refers to an —NH 2 group.
  • Alkylamino refers to an amino group substituted by an alkyl group and “dialkylamino” refers to an amino group substituted by two alkyl groups.
  • aminoalkyl refers to an alkyl group substituted by an amino group.
  • carbonyl refers to >C ⁇ O.
  • hydroxy refers to —OH.
  • mercapto refers to —SH.
  • ureido refers to —NHCONH 2 .
  • sulfonyl refers to >SO 2 .
  • oxy refers to —O—.
  • alkylcarbonylalkenyl refers to an alkenyl group substituted by a carbonyl group which in turn is substituted by an alkyl group.
  • alkylcarbonylalkenyl refers to an alkenyl group substituted by a carbonyl group which in turn is substituted by an alkyl group.
  • carbocyclylalkyl refers to an alkyl moiety substituted by a carbocyclyl group.
  • Example carbocyclylalkyl groups include “aralkyl” (alkyl substituted by aryl) and “cycloalkylalkyl” (alkyl substituted by cycloalkyl).
  • carbocyclylalkenyl refers to an alkenyl moiety substituted by a carbocyclyl group.
  • Example carbocyclylalkenyl groups include “aralkenyl” (alkenyl substituted by aryl) and “cycloalkylalkenyl” (alkenyl substituted by cycloalkyl).
  • carbocyclylalkynyl refers to an alkynyl moiety substituted by a carbocyclyl group.
  • Example carbocyclylalkynyl groups include “aralkynyl” (alkynyl substituted by aryl) and “cycloalkylalkynyl” (alkynyl substituted by cycloalkyl).
  • heterocyclylalkyl refers to an alkyl moiety substituted by a heterocarbocyclyl group.
  • Example heterocyclylalkyl groups include “heteroarylalkyl” (alkyl substituted by heteroaryl) and “heterocycloalkylalkyl” (alkyl substituted by heterocycloalkyl).
  • heterocyclylalkenyl refers to an alkenyl moiety substituted by a heterocyclyl group.
  • Example heterocarbocyclylalkenyl groups include “heteroarylalkenyl”(alkenyl substituted by heteroaryl) and “heterocycloalkylalkenyl” (alkenyl substituted by heterocycloalkyl).
  • heterocyclylalkynyl refers to an alkynyl moiety substituted by a heterocarbocyclyl group.
  • Example heterocyclylalkynyl groups include “heteroarylalkynyl”(alkynyl substituted by heteroaryl) and “heterocycloalkynylalkyl” (alkynyl substituted by heterocycloalkyl).
  • the phrase “5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom” refers to 5- or 6-membered heterocycloalkyl groups or 5- or 6-membered heteroaryl groups, each containing within the 5- or 6-membered ring at least one N atom and optionally additional heteroatoms (e.g., 1, 2 or 3additional heteroatoms selected from N, O and S).
  • Example 6-membered heterocyclic groups containing at least one ring-forming N atom include aromatics such as pyridyl (i.e., pryidinyl), pyrimidinyl, pyrazinyl, triazinyl, and the like.
  • Example 6-membered heterocyclic groups containing at least one ring-forming N atom include non-aromatics such as piperidinyl, piperazinyl, morpholino, and the like.
  • Example 5-membered heterocyclic groups containing at least one ring-forming N atom include aromatics such as imidazolyl, pyrrolyl, pyrrazolyl, trizaolyl, oxoazoyl, thiazolyl and the like.
  • Example 5-membered heterocyclic groups containing at least one ring-forming N atom include non-aromatics such as pyrrolidine, imidazolino, imidazolidino, and the like.
  • the 5- or 6-membered ring can be attached through either a C atom or a heteroatom.
  • the 5- or 6-membered ring can be fused to an aryl or heteroaryl group, meaning that the 5- or 6-membered ring shares a bond with an aryl or heteroaryl group.
  • the fused aryl or heteroaryl group can be a 5- or 6-membered group such as phenyl, naphthyl, pyridyl, pyimidinyl, pyrazinyl, triazinyl, imidazolyl, pyrrolyl, pyrrazolyl, trizaolyl, oxoazoyl, thiazolyl and the like.
  • Example 5- or 6-membered heterocyclic groups containing at least one ring-forming N atom and which are fused to an aryl or heteroaryl group include quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, benzoxazolyl and the like.
  • the 5- or 6-membered heterocyclic group containing at least one ring-forming N atom and which are fused to an aryl or heteroaryl are preferably attached through a C or heteroatom of the 5- or 6-membered heterocyclic group.
  • protecting group refers to a chemical functional group that can be selectively appended to and removed from functionalities, such as hydroxyl groups, amino groups, and carboxyl groups. Protecting groups are usually introduced into a chemical compound to render such functionality inert to chemical reaction conditions to which the compound is exposed. Any of a variety of protecting groups can be employed with the present invention.
  • a protecting group of an amino moiety can be referred to as an “amino protecting group.”
  • Amino protecting groups can have the formulas aryl-SO 2 —, alkyl-SO 2 —, aryl-C( ⁇ O)—, aralkyl-C( ⁇ O)—, alkyl-C( ⁇ O)—, aryl-OC( ⁇ O)—, aralkyl-OC( ⁇ O)—, alkyl-OC( ⁇ O)—, aryl-NHC( ⁇ O)—, alkyl-NHC( ⁇ O)—, and the like, wherein said alkyl, aryl and aralkyl groups may be substituted or unsubstituted.
  • Example amino and guanidino protecting groups can also include t-butyloxycarbonyl (BOC), fluorenylmethoxycarbonyl (Fmoc), benzyloxycarbonyl (Cbz), and a phthalimido group. Further representative protecting groups can be found in T. W. Green and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd. Ed., Wiley & Sons, Inc., New York (1999), which is incorporated herein by reference in its entirety.
  • substituted indicates that at least one hydrogen atom of a chemical group is replaced by a non-hydrogen moiety.
  • substituents include F, Cl, Br, I, C 1 -C 6 alkyl, C 1 -C 6 alkenyl, C 1 -C 6 , alkynyl, haloalkyl, NR E R F , N 3 , NO 2 , CN, CNO, CNS, C( ⁇ O)OR E , R E CO, R E C( ⁇ O)O, R E CONR E , R E R F NCO, ureido, OR E , SR E , SO 2 -alkyl, SO 2 -aryl, and SO 2 —NR E R F , wherein R E and R F are each, independently, H or C 1 -C 6 alkyl.
  • R E and R F may be combined, with the nitrogen to which they are attached, to form a 5 to 7 membered heterocyclic ring, for example pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, and N-methylpiperazinyl.
  • a chemical group herein is “substituted” it may have up to the full valance of substitution, provided the resulting compound is a stable compound or stable structure; for example, a methyl group may be substituted by 1, 2, or 3 substituents, a methylene group may be substituted by 1 or 2 substituents, a phenyl group may be substituted by 1, 2, 3, 4, or 5 substituents, and the like.
  • leaving group refers to any group that can be replaced by a nucleophile upon nucleophilic substitution.
  • Example leaving groups include, halo (F, Cl, Br, I), hydroxyl, alkoxy, mercapto, thioalkoxy, triflate, alkylsulfonyl, substituted alkylsulfonate, arylsulfonate, substituted arylsulfonate, heterocyclosulfonate or trichloroacetimidate.
  • Representative examples include p-(2,4-dinitroanilino)benzenesulfonate, benzenesulfonate, methylsulfonate, p-methylbenzenesulfonate, p-bromobenzenesulfonate, trichloroacetimidate, acyloxy, 2,2,2-trifluoroethanesulfonate, imidazolesulfonyl and 2,4,6-trichlorophenyl.
  • stable compound or “stable structure” refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and preferably capable of formulation into an efficacious therapeutic agent.
  • the present invention is directed only to stable compounds.
  • the compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated.
  • Compounds of the present invention that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C ⁇ N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. Cis and trans geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms.
  • the compounds herein described may have asymmetric centers which result in one enantiomer of a compound of Formula (I) demonstrating superior biological activity over the opposite enantiomer.
  • separation of the racemic material can be achieved by methods known in the art.
  • Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium.
  • phrases “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • the present invention also includes pharmaceutically acceptable salts of the compounds described herein.
  • pharmaceutically acceptable salts refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like
  • organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic,
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
  • Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and in the Journal of Pharmaceutical Science, 66, 2 (1977), the disclosures of each of which are hereby incorporated by reference.
  • the reactions for preparing compounds of the invention can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis.
  • suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected.
  • Preparation of compounds of the invention can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd. Ed., Wiley & Sons, Inc., New York (1999), which is incorporated herein by reference in its entirety.
  • Reactions can be monitored according to any suitable method known in the art.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry
  • chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • Compounds of the invention can be prepared according to methods for preparing aminoboronic acids, esters thereof, and related compounds described in the art, such as in U.S. Pat. Nos. 4,537,773, and in 5,614,649, each of which is incorporated herein by reference in its entirety.
  • the present compounds can be prepared by the sequential coupling of three fragment components (F1, F2, and F3) as described below.
  • Synthesis of compounds of the invention can involve a boron-containing fragment (F1) having a general structure indicated by Formula (A).
  • the boronic ester moiety of F1 can include, for example, a diol ester such as is indicated by the loop connecting oxygen atoms in Formula (A).
  • Stereochemistry at the carbon atom alpha to the boron atom in Formula (A) can be controlled using an asymmetric boronic ester group in the preparation of F1.
  • pinanediol esters of boronic acid can facilitate the preparation or stereochemically pure, or substantially stereochemically pure, F1 fragment.
  • the F1 fragment can be prepared by reacting a compound of Formula (B) (showing a pinanediol boronic ester obtained from (+)-pinanediol) with a strong base (e.g., lithium diisopropylamide or lithium dicyclohexylamide) in the presence of dichloromethane or dibromomethane, followed by addition of a Lewis acid, (e.g., ZnCl 2 , ZnBr 2 , or FeCl 3 ) to yield a compound of Formula (C) (where L is halo) having a newly introduced stereocenter at the carbon alpha to the boron.
  • a strong base e.g., lithium diisopropylamide or lithium dicyclohexylamide
  • a Lewis acid e.g., ZnCl 2 , ZnBr 2 , or FeCl 3
  • the compound of Formula (C) can, in turn, be reacted with an alkali amide (e.g., lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide, and potassium bis(trimethylsilyl)amide) or other nucleophile that effectively inverts the newly formed stereocenter (such as by an SN2 type mechanism) and introduces an amine group (NR 2 ) in place of the leaving group L (e.g., chloro), forming a compound of Formula (D) (where each R can independently be, e.g., alkyl, Si(alkyl) 3 , aryl, or aralkyl).
  • an alkali amide e.g., lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide, and potassium bis(trimethylsilyl)amide
  • NR 2 an amine group in place of the leaving group L (e.g., chloro)
  • the compound of Formula (D) can be further reacted with an agent capable of converting the NR 2 group to NH 2 , or salt thereof, to form an F1 fragment substantially capable of coupling with a further fragment through the amine.
  • a suitable agent for converting the NR 2 group to NH 2 can be a protic acid such as HCl such as when R is a silyl group (e.g., trimethylsilyl).
  • the compound of Formula (B) can also be prepared according to a two step procedure involving reaction of a trialkoxyborane, such as triisopropoxyborane, with (1S, 2S, 3R, 5S)-(+) pinanediol, to give a mono-alkoxy [(1S, 2S, 3R, 5S)-(+) pinanediol]borane intermediate wherein two of the alkoxy groups of the trialkoxy borane are replaced by (1S, 2S, 3R, 5S)-(+) pinanediol.
  • This mixed pinanediol alkoxy borane upon reaction with the appropriate organometallic derivative, e.g.
  • the mid-section of compounds of the present invention can be represented by fragment F2 which couples to fragment F1 by peptide bond formation for form an F2-F1 intermediate.
  • Methods for coupling compounds through peptide bonds, or amide bonds, are well known in the art and described, for example, in The Peptides: Analysis, Synthesis, Biology , Vol. I., eds. Gross, et al., Academic Press, 1979, which is incorporated herein by reference in its entirety.
  • An example F2 fragment is provided in Formula (E) (Pg is an amino protecting group, Z is defined herein). Additionally, protection of the amino group of amino acids using Boc or other amino protecting groups is well known in the art.
  • aza-serines can be prepared generally by the Hoffman Rearrangement (Hoffman's Reaction) using, for example, asparagine where the amide of the asparagine side chain is converted to an amine (which can be subsequently protected). Methods for carrying out Hoffman Rearrangements, such as for amino acids, are known in the art and also provided in the Examples below. Additionally aza-serines can be prepared as disclosed in Zhang, et al. J. Org. Chem., 1997, 62, 6918-6920, which is incorporated herein by reference in its entirety. F2 fragments can be obtained from commercial sources or made by methods known to one skilled in the art.
  • a further fragment (F3) can be coupled to the F2 fragment of the F2-F1 intermediate by any of various means such as by nucleophilic substitution or addition reactions where, for example, F2 contains a nucleophile (e.g., amine) and F3 contains an electrophile (e.g., CO) and optionally a leaving group (e.g., halo, hydroxy, alkoxy, alkylsulfonyl, arylsulfonyl, and the like).
  • Example F3 fragments can have the formula HyCOOH.
  • Coupling of HyCOOH to the F2-F1 intermediate can be carried out according to standard procedures for peptide bond formation to prepare compounds having the formula F3-F2-F1 where the F3and F2 fragments are coupled via an amide bond.
  • Other coupling means are known in the art and are also suitable.
  • F3 fragments can be obtained from commercial sources or made by methods known in the art.
  • the F3-F2-F1 product includes compounds of the invention and can also be derivatized to prepare additional compounds of the invention by routine methods in the art.
  • compounds of the invention where Z is —CH 2 NH 2 can be prepared by removal of an amino protecting group such as benzyloxycarbonyl group (—C( ⁇ O)OCH 2 (C 6 H 5 )) which is attached to one of the nitrogens of the azaserine group (e.g., compounds of Formula (I) where Z is —CH 2 NHR 1 and R 1 is —C( ⁇ O)OCH 2 (C 6 H 5 )).
  • Removal of the benzyloxycarbonyl group can be carried out by treatment with a reducing agent, such as a hydrogenation reagent.
  • the hydrogenation reagent contains H 2 which is optionally used in the presence of a metal catalyst (e.g., Pd/C 10%). Hydrogenation can be further carried out in the presence of a protic acid such as HCl and in a suitable hydrogenation solvent containing, for example, an alcohol (e.g., methanol) and/or an ether solvent (e.g., 1,4-dioxane).
  • a metal catalyst e.g., Pd/C 10%
  • Hydrogenation can be further carried out in the presence of a protic acid such as HCl and in a suitable hydrogenation solvent containing, for example, an alcohol (e.g., methanol) and/or an ether solvent (e.g., 1,4-dioxane).
  • Certain compounds of the invention wherein Z is —CH 2 NHR 1 can be prepared by removal of an R 1 amino protecting group to form the corresponding deprotected amine (such as described above) followed by reaction with a reagent having the formula R 1 X L , (with the exception that R 1 is not H; and X L is a leaving group such as halo or a sulfonic acid derivative; or wherein R 1 and X L taken together represent, for example, a reactive alkyl, carbocyclyl or heterocarbocyclyl isocyanate, or an alkyl, carbocyclyl, heterocarbocyclyl sulphonylisocyanate).
  • boronic esters such as pinanediol esters
  • Hydrolysis conditions can include contacting a boronic ester with excess acid, such as a protic acid like HCl.
  • boronic acids can be esterified by contacting the acid compound (—B(OH) 2 ) with an alcohol such as a diol for sufficient time to produce the corresponding ester.
  • the esterification reaction can be acid or base catalyzed.
  • Step 1 2-(2-methylpropyl)-(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole
  • Step 2 2-[(1S)-1-chloro-3-methylbutyl]-(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole
  • a solution of lithium diisopropylamide was prepared by addition of 10.0 M butyl lithium solution in hexane (25.4 ml, 0.254 mol) to a solution of diisopropylamine (35.7 ml, 0.254 mol) in dry tetrahydrofuran (60 ml), at ⁇ 50° C., and allowing the temperature to rise to ⁇ 30° C.
  • Step 3 N,N-Bis(trimethylsilyl)-(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine
  • Step 4 (1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt
  • Step 1 2-(1-methylethoxy)-(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole
  • Step 2 2-(2-methylpropyl)-(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole
  • Boc-L-threonine (870 mg, 3.97 mmol, 1.2 eq.) was dissolved in DMF dry (30 ml) at r.t. To this solution, TBTU (N,N,N′,N′-tetramethyl-O-(benzotriazol-1-yl)uronium tetrafluoroborate; 1270 mg, 3.97 mmol, 1.2 eq.) was added and the mixture was cooled to 0°to 5° C.
  • NMM N-methylmorpholine, 0.9 ml, 8.27 mmol, 2.5 eq.
  • (1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt of Example A.1 1000 mg, 3.3 mmol, 1 eq.
  • Example G.7 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid, (300 mg, 1 mmol, 1.2 eq.) of Example G.7 was dissolved in DMF dry (25 ml), under nitrogen, and TBTU (318 mg, 1 mmol, 1.2 eq.) was added at r.t. The mixture was cooled at 0°-5° C.
  • Example G.10 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid, (0.41 g, 1.26 mmol, 1.2 eq.) of Example G.10, was dissolved in DMF dry (20 ml), under nitrogen, and TBTU (0.40 g, 1.26 mmol, 1.2 eq.) was added at r.t. The mixture was cooled at 0°-5° C.
  • Step 1 N-tert-butoxycarbonyl-L-asparagine [Commercially available]
  • Step 2 N-[(1,1-dimethylethoxycarbonyl)amino]-L-asparagine, benzyl ester
  • the compound was prepared according to Bioorg. Med. Chem., 6 (1998)1185-1208. N-[(1,1-dimethylethoxycarbonyl)amino]-L-asparagine (20.7 g, 89.1 mmol, 1 eq.), of Step 1, was dissolved in methanol (500 ml) and cesium carbonate (15.97 g, 49 mmol, 0.55 eq.) was added. The solvent was evaporated to give a white solid that was dissolved in N,N-dimethylformamide (200 ml). To the suspension, benzyl bromide (11.6 ml, 98 mmol, 1.1 eq.) was added dropwise and the mixture was stirred overnight.
  • Step 3 3-Amino-2-S-[(1,1-dimethylethoxycarbonyl)amino]-propionic acid, benzyl ester
  • N-[(1,1-dimethylethoxycarbonyl)amino]-L-asparagine, benzyl ester, (2 g, 6.3 mmol, 1 eq.), of Step 2 was dissolved in acetonitrile (80 ml) and water (80 ml). The solution was cooled to 0°-5° C. and iodobenzene diacetate (3 g, 9.3 mmol, 1.5 eq.) was added portionwise. The mixture was stirred at 0° C. for 30′, then at r.t. for 4 h. The organic solvent was removed under vacuum, diethyl ether and HCl 1N were added.
  • Step 1 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(4-methylbenzoylamino)propionic acid, benzyl ester
  • Step 2 (2S)-2-[(1,1-dimethylethoxycarbonyl)amino]-3-(4-methylbenzoylamino)propionic acid
  • Step 1 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid, benzyl ester
  • Hexanoic acid (450 mg, 3.87 mmol, 1.2 eq.) was dissolved in DMF dry (15 ml) and TBTU (1.24 g, 3.87 mmol, 1.2 eq.) was added, the mixture was stirred at r.t. for 20′, then was cooled to 0°-5° C. with ice bath.
  • Step 2 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid
  • Step 1 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(4-fluorosulfonylamino)propionic acid, benzyl ester
  • Step 2 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(4-fluorosulfonylamino)propionic acid
  • Step 1 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(3,4-dimethoxyphenylacetamido)-propionic acid, benzyl ester
  • 3,4-Dimethoxy-phenylacetic acid (720 mg, 3.66 mmol, 1.2 eq.) was dissolved in DMF dry (20 ml) and TBTU (1.17 g, 3.66 mmol, 1.2 eq.) was added, the mixture was stirred at r.t. for 20′, then was cooled to 0°-5° C. with ice bath.
  • 3-amino-2-S-tert-butoxycarbonylamino-propionic acid, benzyl ester (0.9 g, 3.05 mmol, 1 eq.), of Example G.5, and NMM (1.0 ml, 9.15 mmol, 2.5 eq.) were added. The mixture was stirred at 0° C.
  • Step 2 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(3,4-dimethoxyphenylacetamido)-propionic acid
  • Step 1 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid, benzyl ester
  • Step 2 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid
  • G.11.1 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(acetamido)propionic acid.
  • G.11.2 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(9- fluorenylmethyloxycarbamoyl))propionic acid.
  • G.11.3 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(3- pentylureido)propionic acid.
  • G.11.4 2-[(1,1-dimethylethoxycarbonyl)amino]-3- (methanesolfonamido)propionic acid.
  • G.11.5 2-[(1,1-dimethylethoxycarbonyl)amino]-3- [(ethoxycarbonylsuccinyl]-amide)ethyl]-propionic acid.
  • Step 1 N 2 -(tert-butoxycarbonyl)-L-2,3-diaminopropionic acid
  • N-tert-butoxycarbonyl-L-asparagine from step 1 of Example G.5 or commercially available, (8 g, 0.034 mol, 1 eq.) was suspended in ethyl acetate (72 ml), acetonitrile (72 ml) and water (36 ml), and iodobenzenediacetate (13.3 g, 0.041 mol, 1.2 eq.) was added at 5° C. The mixture was stirred at 10°-25° C. for 3-4 h, then a white solid came off. The solid was filtered, washed with diethyl ether and dried under vacuum to give a white powder. Yield 57%. 4 g.
  • Step 2 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(3-benzyloxycarbonylamino)propionic acid
  • N 2 -(tert-Butoxycarbonyl)-L-2,3-diaminopropionic acid, from step 1, (3.8 g, 0.018 mol, 1 eq.) was dissolved in aqueous sodium carbonate 10% (2.2 eq.) at 25° C. and 1,4-dioxane (38 ml).
  • benzylchloroformate (3 ml, 0.020 mol, 1.1 eq.) was added dropwise and the solution was stirred at 25° C. for 3 h.
  • the mixture was poured in water (100 ml) and washed with diethyl ether (100 ml).
  • the intermediate was prepared according to the procedure described in Vederas, J. Am. Chem. Soc., 1985, 107, 7105-7109.
  • 6-Phenyl-pyrazine-2-carboxylic acid (192 mg, 0.96 mmol) and TBTU (310 mg, 0.96 mmol) were dissolved in dry DMF (4 ml), under a nitrogen atmosphere. The reaction mixture was cooled to 0° C. and N-methyl-morpholine (2.29 ml, 2.61 mmol) was added.
  • 2-Pyrazinecarboxylic acid (163 mg, 1.31 mmol, 1.1 eq.) was dissolved in dry DMF (6 ml).
  • TBTU (420 mg, 1.11 mmol, 1.1 eq.) was added to the solution at r.t. under nitrogen. The resulting mixture was stirred for 10 min and then cooled to 0-5° C.
  • Carboxylic acids suitable for preparing compounds of the invention according to, for example, the procedures of Examples H.1 and J.1 can be obtained commercially or prepared according to routine methods or known synthetic procedures.
  • 6-phenyl-pyrazine -2-carboxylic acid and 5-phenyl-pyrazine-2-carboxylic acid were prepared according to the procedure described in Litmanowitsch et al. CH458361.
  • 6-Butyl-pyridine-2-carboxylic acid was prepared via a Reissert-Kaufman type reaction according the procedures described by Honma et al. in J. Med.
  • proteasome inhibitors can modulate, such as induce, apoptosis in a cell.
  • the compounds herein can kill tumor cells by induction of apoptosis.
  • the present compounds can be used to treat cancer, tumors or other proliferative disorders.
  • inhibition of proteasome function by compounds of the invention can inhibit the activation or processing of transcription factor NF- ⁇ B.
  • This protein plays a role in the regulation of genes involved in the immune and inflammatory responses as well as in cell viability.
  • Inhibition of proteasome function can also inhibit the ubiquitination/proteolysis pathway. This pathway catalyzes, inter alia, selective degradation of highly abnormal proteins and short-lived regulatory proteins.
  • compounds of the invention can prevent the degradation of p53 which is typically degraded by the ubiquitin-dependent pathway.
  • the ubiquitination/proteolysis pathway also is involved in the processing of internalized cellular or viral antigens into antigenic peptides that bind to MHC-I molecules.
  • the compounds of the invention can be used to reduce the activity of the cytosolic ATP-ubiquitin-dependent proteolytic system in a number of cell types.
  • the usefulness of such compounds can include therapeutics, such as the treatment of various diseases or disorders associated with proteasome.
  • the methods include administering a therapeutically effective amount of a compound of the invention, or composition thereof, to a mammal, such as a human having a disease or disorder associated with proteasome.
  • therapeutically effective amount refers to an amount sufficient to prevent, alleviate, or ameliorate any phenomenon, such as a cause or symptom, known in the art to be associated with the disease or disorder.
  • Treatable diseases or disorders can be associated with either normal or abnormal activities of proteasome, such as the regulation of apoptosis.
  • proteasome such as the regulation of apoptosis.
  • Numerous diseases or disorders that are associated with proteasome, or that are desirably treated by induction of apoptosis, are known and include, for example, various cancers and tumors including those associated with skin, prostate, colorectal, pancreas, kidney, ovary, mammary, liver, tongue, lung, and smooth muscle tissues.
  • Preferred tumors that can be treated with proteasome inhibitors include, but are not limited to hematological tumors, such as, for example, leukemias, lymphomas, non-Hodgkin lymphoma, myeloma, multiple myeloma, as well as solid tumors such as, for example, colorectal, mammary, prostate, lung, and pancreas tumors.
  • hematological tumors such as, for example, leukemias, lymphomas, non-Hodgkin lymphoma, myeloma, multiple myeloma
  • solid tumors such as, for example, colorectal, mammary, prostate, lung, and pancreas tumors.
  • the proteasome inhibitors can be administered to patients as single agents or in combination with one or more antitumor or anticancer agent and/or radiotherapy.
  • Examples of other anti-tumor or anti-cancer agents which can be advantageously administered concomitantly with a proteasome inhibitor include but are not limited to, adriamycin, daunomycin, methotrexate, vincristin, 6-mercaptopurine, cytosine arabinoside, cyclophosphamide, 5-FU, hexamethylmelamine, carboplatin, cisplatin, idarubycin, paclitaxel, docetaxel, topotecan, irinotecam, gemcitabine, L-PAM, BCNU and VP-16.
  • Methods for determining apoptosis in vitro are well known in the art and kits are available commercially. See for example the Apo-ONETM Homogeneous Caspase-3/7 Assay from Promega Corporation, Madison Wis., USA (Technical Bulletin No. 295, revised 2/02, Promega Corporation).
  • accelerated or enhanced proteolysis that occurs in atrophying muscles, such as is often associated with activation of a nonlysomal ATP-requiring process involving ubiquitin.
  • Accelerated or enhanced proteolysis can be the result of any of numerous causes including sepsis, burns, trauma, cancer, infection, neurodegenerative diseases such as muscular dystrophy, acidosis, or spinal/nerve injuries, corticosteroid use, fever, stress, and starvation.
  • Compounds of the invention can be tested for inhibition of muscle wastage by any various procedures known in the art such as by measuring urinary excretion of modified amino acid 3-methylhistidine (see, e.g., Young, et al., Federation Proc., 1978, 37, 229).
  • Compounds of the present invention can be further used to treat or prevent diseases or disorders associated with activity of NF- ⁇ B including for example, human immunodeficiency virus (HIV) infection and inflammatory disorders resulting from, for example, transplantation rejection, arthritis, infection, inflammatory bowel disease, asthma, osteoporosis, osteoarthritis, psoriasis, restenosis, and autoimmune diseases.
  • HIV human immunodeficiency virus
  • inflammatory disorders resulting from, for example, transplantation rejection, arthritis, infection, inflammatory bowel disease, asthma, osteoporosis, osteoarthritis, psoriasis, restenosis, and autoimmune diseases Accordingly, a process that prevents activation of NF- ⁇ B in patients suffering from such a disease would be therapeutically beneficial. Inhibition of the NF- ⁇ B activity can be measured by using a DNA binding assay such a described in Palombella, et al., Cell, 1994, 78, 773.
  • Proteasome chymotrypsin-like activity of compounds of the invention can be assayed according to the following procedure.
  • HEP Human Erythrocyte Proteasome
  • the concentration of free AMC (aminomethylcoumarin) can be determined on a Perkin Elmer HTS 7000 Plus microplate reader, excitation 370 nM and emission 465 nM.
  • Proteasome activity can be determined under conditions in which substrate hydrolysis increased linearly with time and the change in fluorescence signal is proportional to the concentration of free AMC.
  • bovine ⁇ -chymotrypsin In 96-well microtiter plates bovine ⁇ -chymotrypsin, purchased from Sigma Inc., can be plated at 10 ng/mL (about 2 ⁇ M catalytic sites) in 0.5 M NaCl 50 mM Hepes buffer.
  • a fluorimetric substrate Suc-AAPF-AMC succinyl-Ala-Ala-Pro-Phe-7-amido-4-methylcoumarin
  • Reaction volumes are 100 ⁇ l per well.
  • the concentration of free AMC can be determined on a Perkin Elmer HTS 7000 Plus microplate reader, excitation 370 nM and emission 465 nM.
  • ⁇ -Chymotrypsin activity can be determined under conditions in which substrate hydrolysis increased linearly with time and the change in fluorescence signal was proportional to the concentration of free AMC.
  • IC 50 values are typically defined as the concentration of a compound necessary to produce 50% inhibition of the enzyme's activity. IC 50 values are useful indicators of the activity of a compound for its designated use.
  • the proteasome inhibitors of the invention can be considered active if they have IC 50 values of less than about 1 micromolar for inhibition of human erythrocyte proteasome (HEP).
  • the inhibitors show some specificity for HEP and the ratio of the IC 50 for inhibition of bovine ⁇ -chymotrypsin versus the IC 50 for inhibition of HEP, i.e, IC 50 ( ⁇ -Chymotripsin)/IC 50 (HEP), is greater then about 100.
  • Inhibition of the chymotrypsin-like activity of HEP and of bovine ⁇ -chymotrypsin can be determined by incubating the enzyme with various concentrations of putative inhibitors for 15 minutes at 37° C. (or room temperature for ⁇ -chymotrypsin) prior to the addition of substrate. Each experimental condition can be evaluated in triplicate.
  • Compounds of the present invention are considered active in the above identified assay if their IC 50 values for inhibition of HEP are less than 1000 nanoMolar.
  • compounds of the present invention will have IC 50 values for inhibition of HEP less than 100 nanoMolar. More preferably compounds of the present invention will have IC 50 values for inhibition of HEP less than 10 nanoMolar.
  • the chymotrypsin-like activity of proteasome in Molt-4 cells can be assayed according to the following procedure. A brief description of the method was published previously (Harding et al., J. Immunol., 1995, 155, 1767).
  • Molt-4 cells are washed and resuspended in HEPES-buffered Saline (5.4 mM KCl, 120 mM NaCl, 25 mM Glucose, 1.5 mM MgSO 4 , 1 mM Na pyruvate, 20 mM Hepes) and plated in 96-well microtiter white plates to a final concentration of 6 ⁇ 10 6 cells/mL. Then various 5 ⁇ proteasome inhibitor concentrations (or diluted DMSO for controls), prepared from 250 ⁇ DMSO solutions by diluting 50-fold using HEPES-buffered saline, are added to the plate to a final 1 ⁇ concentration.
  • HEPES-buffered Saline 5.4 mM KCl, 120 mM NaCl, 25 mM Glucose, 1.5 mM MgSO 4 , 1 mM Na pyruvate, 20 mM Hepes
  • a fluorimetric cell permeable substrate (MeOSuc-FLF-AFC) (methoxysuccinyl-Phe-Leu-Phe-7-amido-4-trifluoromethylcoumarin) purchased from Enzyme Systems Products, catalogue number AFC-88, is added to each well at a final concentration of 25 ⁇ M from a stock solution of 20 mM in DMSO. Reaction volumes can be 100 ⁇ l per well.
  • the concentration of free AFC is monitored every 1.5 min for 30 min (22 cycles) on a Polastar Optima, BMG Labtechnologies microplate reader, using an excitation wavelength of 390 nm and emission wavelength of 520 nm.
  • Proteasome activity can be determined under conditions in which substrate hydrolysis increased linearly with time and the change in fluorescent signal is proportional to the concentration of free AFC.
  • EC 50 values are typically defined as the concentration of a compound required to produce an inhibition of the enzyme's activity halfway between the minimum and the maximum response (0% and 85-90% respectively for this assay). EC 50 values are useful indicators of the activity of a compound for its designated use. The compounds of the invention can be considered active if they have an EC 50 of less than about 10 micromolar.
  • Inhibition of chymotrypsin-like activity of proteasome in Molt-4 cells is determined by incubating cells with various concentrations of putative inhibitors for 15 minutes at 37° C. prior to the addition of substrate. Each experimental condition is evaluated in triplicate.
  • Compounds of the present invention are considered active in the above identified assay if their EC 50 values for proteasome inhibition in MOLT-4 are less than 10 microMolar.
  • compounds of the present invention will have EC 50 values for proteasome inhibition in MOLT-4 less than 2 microMolar. More preferably compounds of the present invention will have EC 50 values for proteasome inhibition in MOLT-4 less than 200 nanomolar.
  • the trypsin-like activity of human proteasome can be assayed as described above with the following modifications. Reactions can be carried out in Tris-glycerol buffer (pH 9.5) supplemented with 1 mM 2-mercaptoethanol, and the substrate can be a fluorogenic substrate such as benzyloxycarbonyl-Phe-Arg-AMC (100 ⁇ M).
  • the concentration of free AMC can be determined on a Fluoroskan II spectrofluorimeter with an excitation filter of 390 nm and an emission filter of 460 nm.
  • Protease activity can be determined under conditions in which substrate hydrolysis increases linearly with time and the change in fluorescence is proportional to the concentration of free AMC.
  • the effect of inhibitors on the unweighting atrophy of the soleus muscle in juvenile rats can be determined by, for example, the procedures described in Tischler, Metabolism, 1990, 39, 756.
  • juvenile female Sprague-Dawley rats 80-90 g
  • the animal's hind limbs can be elevated above the floor of the cage with each animal housed individually.
  • Animals can have free access to food and water, and can be weighed at the time of suspension and at time of termination. During the suspension period the animals can be checked daily to ensure that their toes are not touching the floor of the cage, and that there is no swelling of the tail due to the cast.
  • Each experiment can begin with the suspension of 20 rats which are randomly divided into 4 groups of 5 animals each.
  • Group A can be suspended for 2 days, providing baseline data to approximate the soleus muscle size in other animals suspended for longer times. Average body weights for the groups at the outset of the study can be compared and used as a correction factor for body size differences.
  • Group B can be a second control group which has the soleus of one limb treated with an aqueous solution of mersalyl after two days of unweighting, to demonstrate the ability to slow muscle atrophy during unweighting, for each group of animals.
  • an aqueous solution of mersalyl 200 nM; 4 ⁇ L/100 g initial body wt
  • the contralateral muscle can be injected with a similar volume of 0.9% saline (“Vehicle”).
  • the animals can be maintained under Innovar-vet (10 ⁇ L/100 g body wt) tranquilization during the in situ injection procedure. After the injections, the animals can be suspended for an additional 24 hours and the soleus can be removed. Groups C and D for each experiment can be used for testing each of two different embodiments of the disclosed compounds. Animals can be treated as in group B, except that 1 mM proteasome inhibitor, contained in dimethysulfoxide (DMSO), can be injected into the soleus of one leg and DMSO only into the contralateral soleus. Thus each experiment consists of two control groups and the testing of proteasome inhibitors of the invention. The completion of five such experiments with different pairs of inhibitors provides for an “n” value of 10 for testing each inhibitor and each can be tested in two different shipments of animals.
  • DMSO dimethysulfoxide
  • the soleus can be excised, trimmed of fat and connective tissue, and carefully weighed.
  • the muscle can then homogenized in 10% trichloroacetic acid (TCA) and the precipitated protein pelleted by centrifugation.
  • TCA trichloroacetic acid
  • the pellet can then be washed once with 10% TCA and once with ethanol:ether (1:1).
  • the final pellet can be solubilized in 4 ml of 1N sodium hydroxide.
  • the sample can be then analyzed for protein content by the biuret procedure, using albumin as a standard.
  • the effect of inhibitors on total muscle protein content can be examined primarily by paired comparison with the untreated contralateral muscle.
  • the ratio of contents can be calculated and then analyzed statistically by analysis of variance (“ANOVA”).
  • ANOVA analysis of variance
  • the left leg can always be the treated leg so that the protein content ratios can be compared to the non-treated control animals as well. In this way, a significant difference can be shown by comparing the protein content of the two legs, as well as the relative effectiveness of the tested inhibitors.
  • a paired student test can also be performed for the effect of each separate treatment.
  • the non-treated control data also provide an estimate of protein content of day 2. This allows approximation of the protein changes over the 24 hours of treatment for each of the Groups B, C, and D.
  • Each experiment can consist of 10 animals with groups of 5 animals being tested with one of the inhibitors for its effect on protein synthesis. Control animals are not needed for this aspect of the study as the contralateral DMSO-treated muscle serves as the paired control for the inhibitor-treated muscle. Each group can be injected as described for groups C and D in part 1. Twenty-four hours after the in situ treatment the fractional rate of protein synthesis can be analyzed in both soleus muscles. Each muscle can be injected with a 0.9% saline solution (3.5 ⁇ l/100 g final body wt) containing 3 H-phenylalanine (50 mM; 1 ⁇ Ci/ m l). Fifteen minutes later the middle two-thirds of the muscle can be excised and the muscle can be processed as described below.
  • the muscle can be first washed for 10 minutes in 0.84% saline containing 0.5 mM cycloheximide, to terminate protein synthesis, and 20 mM cycloleucine, to trap phenylalanine in the cell.
  • the muscle can then be homogenized in 2.5 mL of ice-cold 2% perchloric acid.
  • the precipitated protein can be pelleted by centrifugation.
  • One aliquot of the supernatant can be taken for liquid scintillation counting and another aliquot can be processed for conversion of phenylalanine to phenethylamine to determine the soluble phenylalanine concentration fluorometrically. See, e.g., Garlick, et al., Biochem.
  • the specific activity of phenylalanine in the muscle protein can be determined after hydrolyzing the protein by heating in 6N HCl. The amino acids released can be solubilized in buffer. One aliquot can be taken for scintillation counting and another for analysis of phenylalanine as for the supernatant fraction. The fractional rate of protein synthesis can be calculated as: protein specific activity/intracellular specific activity x time.
  • Analyses of protein synthesis can be on a paired basis for each inhibitor. Student paired t test comparisons of the contralateral muscles can determine whether there is any effect of the inhibitor on protein synthesis. Protein breakdown can be calculated approximately as the fractional rate of protein synthesis (from part 2) plus the fractional rate of protein accretion (from part 1), where protein loss yields a negative value for protein accretion.
  • proteasome inhibitors used for in vivo studies can be formulated in an appropriate medium for intravenous (iv) or oral (po) administration.
  • iv intravenous
  • po oral
  • the compounds can be administered dissolved in 0.9% NaCl, or in mixtures of 0.9% NaCl, solutol HS15 and dimethylsulfoxide, for example in the ratio 87:10:3 (v:v:v), respectively.
  • human and murine tumor cell lines of different histological origin can be used to test the antitumor activity of the compounds of the invention: H460 (human, lung), A2780 (human, ovary), PC-3 (human, prostate), LoVo (human, colon), HCT116 (human, colon), BXPC3 (human, pancreatic), PANC-1 (human, pancreatic), MX-1 (human, mammary), MOLT (human, leukemia), multiple myeloma (human, myeloma), YC8 (murine, lymphoma), L1210 (murine, leukemia), 3LL (murine, lung).
  • mice 5-6 Weeks immunocompetent or immunodeprived mice are purchased from commercial sources, for example from Harlan (Correzzana, Mi Italy). CD1 nu/nu mice are maintained under sterile conditions; sterilized cages, bedding, food and acidified water are used.
  • Solid tumor models of different hystotype can be transplanted subcutaneously (sc.) into the axillary region of immunocompetent mice (murine models) or in immunodeprived mice (human models).
  • Human tumor cell lines originally obtained from ATCC, can be adapted to grow “in vivo” as solid tumor from “in vitro culture”.
  • Hematological human or murine tumor models can be transplanted into different sites (iv, ip, ic or sc) in immunocompetent mice (murine tumors) or in immunodeprived mice (human leukemia, lymphoma and myeloma models), according to their highest tumor take.
  • mice bearing solid (staged) or hematological tumors are randomized in experimental groups (10 mice/group). For solid tumors, an average tumor weight of 80-100 mg for each group is considered to start the treatment; mice with the smallest and largest tumors are discarded.
  • mice are randomly assigned to the drug treatment and to the control group. Animals can be treated iv or orally, depending on the oral bioavailability with the compounds following different treatment schedules: iv weekly or twice weekly, or by daily oral administration.
  • drug treatment can begin when the tumor size ranges between 80-100 mg after tumor transplantation (Day 0).
  • the compounds can be administered in a volume of 10 mL/Kg body weight/mouse in the appropriate solvent.
  • TWI % percentage of primary tumor growth inhibition in comparison with vehicle treated control groups
  • RTWI % Relative tumor growth inhibition in case of staged tumors
  • the antitumor activity can be determined as tumor weight inhibition (TWI %), which is calculated according to the formula:
  • TWI ⁇ ⁇ % 100 - mean ⁇ ⁇ TW ⁇ ⁇ treated mean ⁇ ⁇ TW ⁇ ⁇ controls ⁇ 100
  • the RTWI % (relative percentage of primary tumor growth inhibition in comparison with vehicle treated control groups) is evaluated one week after the last drug treatment, according to the following formula:
  • the Percent of Tumor Regression can be calculated as regressions in terms of relative tumor weight, determined as tumor weight at given day divided by initial tumor weight at the outset the experiment.
  • the antitumor activity can be determined as percentage increase of the median survival time of mice expressed as the ratio (T/C %) of the median survival time of the treated group (T) to that of the control group (C). Animals which are tumour-free at the end of the experiment (60 days after transplantation) are excluded from the calculation and considered as long term survivors (LTS).
  • Toxicity can be evaluated daily on the basis of the gross autopsy findings and the weight loss. Mice are considered to have died of toxicity when death occurs before the death of vehicle treated control animals, or when significant body weight loss (>20%), and/or spleen and liver size reduction are observed.
  • the BWC % (Body weight change %) is assessed as follow: 100 ⁇ (mean body weight of mice at given day/mean body weight at start of treatment) ⁇ 100. This value is determined one week after the last treatment with the test compound.
  • the IC 50 values measuring in vitro viability of cells in the presence of test compounds can be determined according to the following procedure. Cells can be seeded in 96-well plates at varying densities and then assayed using the Calcein-AM viability assay after 24 hours to determine the optimal final density for each cell type. Cells can then be seeded in 96-well plates at the determined density in 100 ⁇ L of an appropriate cell media known to one skilled in the art.
  • Serial dilutions of test compounds can be made so that the concentrations are twice the desired concentration to be evaluated.
  • 100 ⁇ L of the dilution is then added to the cells plated in 100 ⁇ L of media, a final concentration of, for example, 0, 11.7, 46.9, 187.5, 375, and 750 nM can be obtained.
  • Compounds can be added to the plates three to four hours after seeding the cells, then the plates can be incubated at 37° C. for the desired time point (e.g., one, two, or three days).
  • Calcein-AM viability assays can be conducted at the desired time points as follows. Media can be aspirated using a manifold and metal plate to leave approximately 50 ⁇ L/well. The wells can be washed three times with 200 ⁇ L DPBS, aspirating each time with the manifold to leave 50 ⁇ L/well. A 8 ⁇ M solution of Calcein-AM in DPBS can be prepared and 150 ⁇ L can be added to each well. The plates can then be incubated at 37° C. for 30 minutes. After incubation, calcein can be aspirated with the manifold and cells can be washed with 200 ⁇ L DPBS as before. After final aspiration, fluorescence can be measured using a Cytofluor 2300 fluorescence plate reader. Negative controls can contain media and no cells, and experiments can be conducted in triplicate.
  • K i dissociation constants for the equilibrium established when proteasome and test compound interact to form a complex.
  • the reactions can be carried out using SDS-activated 20S proteasome from rabbit muscle, and the proteasome substrate can be Suc-LLVY-AMC.
  • Compounds of the invention can be tested for inhibiting the activity of NF- ⁇ B by carrying out the assay described in Palombella, et al., Cell, 1994, 78, 773).
  • MG63 osteocarcinoma cells can be stimulated by treatment with TNF- ⁇ for designated times.
  • Whole cell extracts can be prepared and analyzed by electrophoretic mobility shift assays using the PRDII probe from the human IFN- ⁇ gene promoter.
  • the compounds of Formula (I) can be administered in the form of pharmaceutical compositions.
  • These compositions can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal, and can be prepared in a manner well known in the pharmaceutical art.
  • compositions which contain, as the active ingredient, one or more of the compounds of Formula (I) above in combination with one or more pharmaceutically acceptable carriers.
  • the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose.
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
  • the compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
  • compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 100 mg, more usually about 10 to about 30 mg, of the active ingredient.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • the active compound can be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 500 mg of the active ingredient of the present invention.
  • the tablets or pills of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
  • liquid forms in which the compounds and compositions of the present invention can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions in can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
  • compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. An amount adequate to accomplish this is referred to as “therapeutically effective amount.” Effective doses will depend on the disease condition being treated as well as by the judgement of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
  • compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the compound preparations typically will be between 3and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
  • the therapeutic dosage of the compounds of the present invention can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician.
  • the proportion or concentration of a compound of the invention in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration.
  • the compounds of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 ⁇ g/kg to about 1 g/kg of body weight per day.
  • the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day.
  • the dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • kits useful for example, in the treatment or prevention of inflammatory diseases, which comprise one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I).
  • kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art.
  • Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.

Abstract

The present invention provides boronic acid compounds, boronic esters, and compositions thereof that can modulate apoptosis such as by inhibition of proteasome activity. The compounds and compositions can be used in methods of inducing apoptosis and treating diseases such as cancer and other disorders associated directly or indirectly with proteasome activity.

Description

CROSS REFERENCE TO RELATED APPLICATIONS
This application is a continuation of U.S. application Ser. No. 11/351,193, filed Feb. 9, 2006, which claims the benefit of U.S. Provisional Application No. 60/652,370, filed Feb. 11, 2005, the entireties of which are incorporated herein.
FIELD OF THE INVENTION
The present invention relates to boronic acid and boronic ester compounds useful as proteasome inhibitors and modulation of apoptosis.
BACKGROUND OF THE INVENTION
The proteasome, (also referred to as multicatalytic protease (MCP), multicatalytic proteinase, multicatalytic proteinase complex, multicatalytic endopeptidase complex, 20S, 26S, or ingensin) is a large, multiprotein complex present in both the cytoplasm and the nucleus of all eukaryotic cells. It is a highly conserved cellular structure that is responsible for the ATP-dependent proteolysis of most cellular proteins (Tanaka, Biochem Biophy. Res. Commun., 1998, 247, 537). The 26S proteasome consists of a 20S core catalytic complex that is capped at each end by a 19S regulatory subunit. The archaebacterial 20S proteasome contains fourteen copies of two distinct types of subunits, α and β, which form a cylindrical structure consisting of four stacked rings. The top and bottom rings contain seven α-subunits each, while the inner rings contain seven β-subunits. The more complex eukaryotic 20S proteasome is composed of about 15 distinct 20-30 kDa subunits and is characterized by three major activities with respect to peptide substrates. For example, the proteasome displays tryptic-, chymotryptic-, and peptidylglutamyl peptide-hydrolytic activities (Rivett, Biochem. J., 1993, 291, 1 and Orlowski, Biochemistry, 1990, 29, 10289). Further, the proteasome has a unique active site mechanism which is believed to utilize a threonine residue as the catalytic nucleophile (Seemuller, et al., Science, 1995, 268, 579).
The 26S proteasome is able to degrade proteins that have been marked by the addition of ubiquitin molecules. Typically, ubiquitin is attached to the 1-amino groups of lysines in a multistep process utilizing ATP and E1 (ubiquitin activating) and E2 (ubiquitin-conjugating) enzymes. Multi-ubiquitinated substrate proteins are recognized by the 26S proteasome and are degraded. The multi-ubiquitin chains are generally released from the complex and ubiquitin is recycled (Goldberg, et al., Nature, 1992, 357, 375).
Numerous regulatory proteins are substrates for ubiquitin dependent proteolysis. Many of these proteins function as regulators of physiological as well as pathophysiological cellular processes. Alterations in proteasome activity have been implicated in a number of pathologies including neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, as well as occlusion/ischaemia reperfusion injuries, and aging of the central nervous system.
The ubiquitin-proteasome pathway also plays a role in neoplastic growth. The regulated degradation of proteins such as cyclins, CDK2 inhibitors, and tumor suppressors is believed to be important in cell cycle progression and mitosis. A known substrate of the proteasome is the tumor suppressor p53 which is involved in several cellular processes (see, e.g., Ko, L. J. Genes Dev., 1996, 10, 1054). Tumor suppressor p53 has been shown to induce apoptosis in several haematopoietic cell lines (Oren, M., Semin. Cancer Biol., 1994, 5, 221). Induction of p53 leads to cell growth arrest in the G1 phase of the cell cycle as well as cell death by apoptosis. Tumor suppressor p53 degradation is known to be carried out via the ubiquitin-proteasome pathway, and disrupting p53 degradation by inhibition of the proteasome is a possible mode of inducing apoptosis.
The proteasome is also required for activation of the transcription factor NF-κB by degradation of its inhibitory protein, IκB (Palombella, et al., Cell, 1994, 78, 773). NF-κB has a role in maintaining cell viability through the transcription of inhibitors of apoptosis. Blockade of NF-κB activity has been demonstrated to make cells more susceptible to apoptosis.
Several inhibitors of the proteolytic activity of the proteasome have been reported. See, for example, Kisselev, et al., Chemistry & Biology, 2001, 8, 739. Lactacystin is a Streptomyces metabolite that specifically inhibits the proteolytic activity of the proteasome complex (Fenteany, et al., Science, 1995, 268, 726). This molecule is capable of inhibiting the proliferation of several cell types (Fenteany, et al., Proc. Natl. Acad. Sci. USA, 1994, 91, 3358). It has been shown that lactacystin binds irreversibly, through its β-lactone moiety, to a threonine residue located at the amino terminus of the β-subunit of the proteasome.
Peptide aldehydes have been reported to inhibit the chymotrypsin-like activity associated with the proteasome (Vinitsky, et al., Biochemistry, 1992, 31, 9421; Tsubuki, et al., Biochem. Biophys. Res. Commun., 1993, 196, 1195; and Rock, et al., Cell, 1994, 78, 761). Dipeptidyl aldehyde inhibitors that have IC50 values in the 10-100 nM range in vitro (Iqbal, M., et al., J. Med. Chem., 1995, 38, 2276) have also been reported. A series of similarly potent in vitro inhibitors from α.-ketocarbonyl and boronic ester derived dipeptides has also been reported (Iqbal, et al., Bioorg. Med. Chem. Lett., 1996, 6, 287, U.S. Pat. Nos. 5,614,649; 5,830,870; 5,990,083; 6,096,778; 6,310,057; U.S. Pat. App. Pub. No. 2001/0012854, and WO 99/30707).
N-terminal peptidyl boronic ester and acid compounds have been reported previously (U.S. Pat. Nos. 4,499,082 and 4,537,773; WO 91/13904; Kettner, et al., J. Biol. Chem., 1984, 259(24), 15106). These compounds are reported to be inhibitors of certain proteolytic enzymes. N-terminal tri-peptide boronic ester and acid compounds have been shown to inhibit the growth of cancer cells (U.S. Pat. No. 5,106,948). A broad class of N-terminal tri-peptide boronic ester and acid compounds and analogs thereof has been shown to inhibit renin (U.S. Pat. No. 5,169,841).
Various inhibitors of the peptidase activities of the proteasome have also been reported. See, e.g., Dick, et al., Biochemistry, 1991, 30, 2725; Goldberg, et al., Nature, 1992, 357, 375; Goldberg, Eur. J. Biochem., 1992, 203, 9; Orlowski, Biochemistry, 1990, 29, 10289; Rivett, et al., Archs. Biochem. Biophys., 1989, 218, 1; Rivett, et al., J. Biol. Chem., 1989, 264, 12215; Tanaka, et al., New Biol., 1992, 4, 1; Murakami, et al., Proc. Natl. Acad. Sci. USA, 1986, 83, 7588; Li et al., Biochemistry, 1991, 30, 9709; Goldberg, Eur. J. Biochem., 1992, 203, 9; and Aoyagi, et al., Proteases and Biological Control, Cold Spring Harbor Laboratory Press (1975), pp. 429-454.
Stein et al., U.S. patent application Ser. No. 08/212,909, filed Mar. 15, 1994, report peptide aldehydes useful for reducing in an animal both the rate of loss of muscle mass and the rate of intracellular protein breakdown. The compounds are also said to reduce the rate of degradation of p53 protein in an animal. Palombella, et al., WO 95/25533, report the use of peptide aldehydes to reduce the cellular content and activity of NF-κB in an animal by contacting cells of the animal with a peptide aldehyde inhibitor of proteasome function or ubiquitin conjugation. Goldberg and Rock, WO 94/17816, report the use of proteasome inhibitors to inhibit MHC-I antigen presentation. Stein, et al., U.S. Pat. No. 5,693,617 report peptidyl aldehyde compounds as proteasome inhibitors useful for reducing the rate of degradation of protein in an animal. Inhibition of the 26S and 20S proteasome by indanone derivatives and a method for inhibiting cell proliferation using indanone derivatives are reported by Lum et al., U.S. Pat. No. 5,834,487. Alpha-ketoamide compounds useful for treating disorders mediated by 20S proteasome in mammals are reported in Wang et al., U.S. Pat. No. 6,075,150. France, et al., WO 00/64863, report the use of 2,4-diamino-3-hydroxycarboxylic acid derivatives as proteasome inhibitors. Carboxylic acid derivatives as proteasome inhibitors are reported by Yamaguchi et al., EP 1166781. Ditzel, et al., EP 0 995 757 report bivalent inhibitors of the proteasome. 2-Aminobenzylstatine derivatives that inhibit non-covalently the chymotrypsin-like activity of the 20S proteasome have been reported by Garcia-Echeverria, et al., Bioorg. Med. Chem. Lett., 2001, 11, 1317.
Some further proteasome inhibitors can contain boron moieties. For example, Drexler et al., WO 00/64467, report a method of selectively inducing apoptosis in activated endothelial cells or leukemic cells having a high expression level of c-myc by using tetrapeptidic boronate containing proteasome inhibitors. Furet et al., WO 02/096933 report 2-[[N-(2-amino-3-(heteroaryl or aryl)propionyl)aminoacyl]amino]alkylboronic acids and esters for the therapeutic treatment of proliferative diseases in warm-blooded animals. U.S. Pat. Nos. 6,083,903; 6,297,217; 5,780,454; 6,066,730; 6,297,217; 6,548,668; U.S. Patent Application Pub. No. 2002/0173488; and WO 96/13266 report boronic ester and acid compounds and a method for reducing the rate of degradation of proteins. A method for inhibiting viral replication using certain boronic acids and esters is also reported in U.S. Pat. No. 6,465,433and WO 01/02424. Pharmaceutically acceptable compositions of boronic acids and novel boronic acid anhydrides and boronate ester compounds are reported by Plamondon, et al., U.S. Patent Application Pub. No. 2002/0188100. A series of di- and tripeptidyl boronic acids are shown to be inhibitors of 20S and 26S proteasome in Gardner, et al., Biochem. J., 2000, 346, 447.
Other boron-containing peptidyl and related compounds are reported in U.S. Pat. Nos. 5,250,720; 5,242,904; 5,187,157; 5,159,060; 5,106,948; 4,963,655; 4,499,082; and WO 89/09225, WO/98/17679, WO 98/22496, WO 00/66557, WO 02/059130, WO 03/15706, WO 03/59898, WO 96/12499, WO 95/20603, WO 95/09838, WO 94/25051, WO 94/25049, WO 94/04653, WO 02/08187, EP 632026, and EP 354522. U.S. patent application Ser. Nos. 10/918,664 and 10/918,610, the disclosures of each of which are incorporated herein by reference in their entireties, further report additional boron-containing peptidyl-like proteasome inhibitors.
A great interest exists, as evidenced by the above references, in drugs which can modulate proteasome activity. For example, molecules capable of inhibiting proteasome activity can arrest or delay cancer progression by interfering with the ordered degradation of cell cycle proteins or tumor suppressors. Accordingly, there is an ongoing need for new and/or improved inhibitors of proteasome.
SUMMARY OF THE INVENTION
The present invention is directed to novel boronic acid and boronic ester compounds useful as proteasome inhibitors and modulation of apoptosis. The subject invention also comprises methods for inhibition of multicatalytic protease (“MCP”) associated with certain disorders, including the treatment of muscle wasting disorders.
In one embodiment are provided compounds having Formula (I):
Figure US08283367-20121009-C00001

wherein constituent members are defined infra, as well as preferred constituent members.
In another embodiments, the present invention provides a compound which is a boronic anhydride of a compound of Formula (I), such as a cyclic boronic anhydride.
In another embodiment the present invention provides a pharmaceutical composition comprising a compound of Formula (I) and a pharmaceutically acceptable carrier.
In another embodiment the present invention provides a method of inhibiting activity of proteasome comprising contacting a compound of Formula (I) with the proteasome.
In another embodiment the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to the cancer a therapeutically effective amount of a compound of Formula (I).
In another embodiment the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to the cancer a therapeutically effective amount of a compound of Formula (I), and wherein the cancer is selected from skin, prostate, colorectal, pancreas, kidney, ovary, mammary, liver, tongue, lung, and smooth muscle tissue.
In another embodiment the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to the cancer a therapeutically effective amount of a compound of Formula (I), and wherein said cancer is selected from leukemia, lymphoma, non-Hodgkin lymphoma, myeloma, and multiple myeloma.
In another embodiment the present invention provides a method of treating cancer comprising administering to a mammal having or predisposed to the cancer a therapeutically effective amount of a compound of Formula (I) in combination with one or more antitumor or anticancer agent and/or radiotherapy.
In another embodiment the present invention provides a method of inhibiting activity of transcription factor NF-κB comprising contacting IκB, the inhibitor of transcription factor NF-κB, with a compound of Formula (I).
In another embodiment, the present invention provides a compound of Formula (I) for use in therapy.
In another embodiment, the present invention provides use of a compound of Formula (I) for the manufacture of a medicament for the treatment of cancer.
These and other features of the compounds will be set forth in expanded form as the disclosure continues.
DETAILED DESCRIPTION
The present invention provides, inter alia, compounds that can inhibit proteasome activity and be used for the treatment of diseases or disorders related to proteasome activity. Compounds of the invention include compounds of Formula (I):
Figure US08283367-20121009-C00002

or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3 or —CH2NR1aR1;
Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R4;
R1 is H, C1-10 alkyl, carbocyclyl, heterocyclyl, C1-10 alkyl-C(═O)—, C2-10 alkenyl-C(═O)—, C2-10 alkynyl-C(═O)—, carbocyclyl-C(═O)—, heterocyclyl-C(═O)—, carbocyclylalkyl-C(═O)—, heterocyclylalkyl-C(═O)—, C1-10 alkyl-S(═O)2—, carbocyclyl-S(═O)2—, heterocyclyl-S(═O)2—, carbocyclylalkyl-S(═O)2—, heterocyclylalkyl-S(═O)2—, C1-C10 alkyl-NHC(═O)—, carbocyclyl-NHC(═O)—, heterocyclyl-NHC(═O)—, carbocyclylalkyl-NHC(═O)—, heterocyclylalkyl-NHC(═O)—, C1-C10 alkyl-OC(═O)—, carbocyclyl-OC(═O)—, heterocyclyl-OC(═O)—, carbocyclylalkyl-OC(═O)—, heterocyclylalkyl-OC(═O)—, C1-10 alkyl-NH—C(═O)—NHS(═O)2—, carbocyclyl-NH—C(═O)—NHS(═O)2—, heterocyclyl-NH—C(═O)—NHS(═O)2—, C1-10 alkyl-S(═O)2—NH—C(═O)—, carbocyclyl-S(═O)2—NH—C(═O)—, heterocyclyl-S(═O)2—NH—C(═O)—, or an amino protecting group; wherein R1 is optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3;
R1a is H. Alternatively, R1a and R1 together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocyclyl group optionally substituted with 1, 2, or 3 R3;
R2 is, independently, H or C1-6 alkyl;
alternatively, two R2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group;
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
R4 is, independently, selected from C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, —OR4a, —SR4a, —CN, halo, haloalkyl, —NH2, —NH(alkyl), —N(alkyl)2, —NHC(═O)O-alkyl, —NHC(═O)alkyl, —COOH, —C(═O)O-alkyl, —C(═O)alkyl, —C(O)H, —S(═O)-alkyl, —S(═O)2-alkyl, —S(═O)-aryl, —S(═O)2-aryl, carbocyclyl optionally substituted with 1, 2 or 3 R5, and heterocyclyl optionally substituted with 1, 2 or 3 R5;
R4a is H, C1-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, carbocyclyl or heterocyclyl;
R5 is, independently, selected from C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
with the proviso that when Z is —CH(OH)CH3and Q is
Figure US08283367-20121009-C00003

then Hy is other than
Figure US08283367-20121009-C00004
In some embodiments, Q is boronic acid (B(OH)2) or a cyclic boronic ester wherein said cyclic boronic ester contains from 6 to 10 carbon atoms and contains at least one cycloalkyl moiety.
In some embodiments, Q is B(OH)2 or pinanediol boronic ester.
In some embodiments, Q is pinanediol boronic ester.
In some embodiments, Z is —CH(OH)CH3.
In some embodiments, Z is —CH2NR1aR1.
In some embodiments, Z is —CH2NHR1.
In some embodiments, Z is —CH2NHR1 and R1 is carbocyclyl-C(═O)— or carbocyclyl-S(═O)2—, each optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3.
In some embodiments, Z is —CH2NHR1 and R1 is aryl-C(═O)— or aryl-S(═O)2—, each optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, F, Cl, Br, I, C1-4 haloalkyl, carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3.
In some embodiments, Z is —CH2NHR1 and R1 is phenyl-C(═O)— or phenyl-S(═O)2—, each optionally substituted with C1-4 alkyl, F, Cl, Br, I, or aryl.
In some embodiments, R1 is aryl-C(═O)— or aryl-S(═O)2—, each optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, F, Cl, Br, I, C1-4 haloalkyl, carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3.
In some embodiments, R1 is aryl-C(═O)— optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, F, Cl, Br, I, and C1-4 haloalkyl.
In some embodiments, R1 is phenyl-C(═O)— optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, F, Cl, Br, I, and C1-4 haloalkyl.
In some embodiments, R1 is —CO-(4-methylphenyl).
In some embodiments, R1 is optionally substituted with 1 or 2 substituents.
In some embodiments, R1 is optionally substituted with 1 substituent.
In some embodiments, R1 is substituted with 1 substituent.
In some embodiments, R1 is substituted with C1-6 alkyl.
In some embodiments, R1 is substituted with methyl.
In some embodiments, Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 R4.
In some embodiments, Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, thiazolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, or indolyl, each optionally substituted by 1, 2 or 3 R4.
In some embodiments, Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, thiazolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, or indolyl, each optionally substituted by 1 or 2 C1-8 alkyl, carbocyclyl optionally substituted with 1, 2 or 3 R5, or heterocyclyl optionally substituted with 1, 2 or 3 R5.
In some embodiments, Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, thiazolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, or indolyl, each optionally substituted by 1 or 2 methyl, ethyl, propyl, butyl, aryl optionally substituted with 1, 2 or 3 R5, or heteroaryl optionally substituted with 1, 2 or 3 R5.
In some embodiments, Hy is pyridyl, pyrimidinyl, pyrazinyl, thiazolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, or indolyl, each optionally substituted by 1 or 2 methyl, ethyl, propyl, butyl, aryl optionally substituted with 1, 2 or 3 R5, or heteroaryl optionally substituted with 1, 2 or 3 R5.
In some embodiments, Hy is pyrazine substituted by at least 1 or 2 R4, unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R4, pyridin-4-yl optionally substituted by 1 or 2 R4, N-oxo-pyridinyl optionally substituted by 1 or 2 R4, pyrimidinyl optionally substituted by 1 or 2 R4, imidazolyl optionally substituted by 1 or 2 R4, thiazolyl optionally substituted by 1 or 2 R4, oxazolyl optionally substituted by 1 or 2 R4, pyrrolyl optionally substituted by 1 or 2 R4, pyrazolyl optionally substituted by 1 or 2 R4, quinolinyl optionally substituted by 1 or 2 R4, isoquinolinyl optionally substituted by 1 or 2 R4, quinoxalinyl optionally substituted by 1 or 2 R4, indolyl optionally substituted by 1 or 2 R4, quinazolinyl optionally substituted by 1 or 2 R4, benzoimidazolyl optionally substituted by 1 or 2 R4, benzothiazolyl optionally substituted by 1 or 2 R4, or benzoxazolyl optionally substituted by 1 or 2 R4.
In some embodiments, Hy is pyrazine substituted by at least 1 or 2 R4, unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R4, pyridin-4-yl optionally substituted by 1 or 2 R4, N-oxo-pyridinyl optionally substituted by 1 or 2 R4, pyrimidinyl optionally substituted by 1 or 2 R4, imidazolyl optionally substituted by 1 or 2 R4, thiazolyl optionally substituted by 1 or 2 R4, oxazolyl optionally substituted by 1 or 2 R4, pyrrolyl optionally substituted by 1 or 2 R4, pyrazolyl optionally substituted by 1 or 2 R4, quinolinyl optionally substituted by 1 or 2 R4, isoquinolinyl optionally substituted by 1 or 2 R4, quinoxalinyl optionally substituted by 1 or 2 R4, indolyl optionally substituted by 1 or 2 R4, quinazolinyl optionally substituted by 1 or 2 R4, benzoimidazolyl optionally substituted by 1 or 2 R4, benzothiazolyl optionally substituted by 1 or 2 R4, or benzoxazolyl optionally substituted by 1 or 2 R4, wherein R4 is C1-6 alkyl, aryl or heterocyclyl.
In some embodiments, Hy is pyrazine substituted by at least 1 or 2 R4, unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R4, pyridin-4-yl optionally substituted by 1 or 2 R4, N-oxo-pyridinyl optionally substituted by 1 or 2 R4, pyrimidinyl optionally substituted by 1 or 2 R4, imidazolyl optionally substituted by 1 or 2 R4, thiazolyl optionally substituted by 1 or 2 R4, pyrazolyl optionally substituted by 1 or 2 R4, quinolinyl optionally substituted by 1 or 2 R4, isoquinolinyl optionally substituted by 1 or 2 R4, quinoxalinyl optionally substituted by 1 or 2 R4, or indolyl optionally substituted by 1 or 2 R4.
In some embodiments, Hy is pyrazine substituted by at least 1 or 2 R4, unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R4, pyridin-4-yl optionally substituted by 1 or 2 R4, N-oxo-pyridinyl optionally substituted by 1 or 2 R4, pyrimidinyl optionally substituted by 1 or 2 R4, imidazolyl optionally substituted by 1 or 2 R4, thiazolyl optionally substituted by 1 or 2 R4, pyrazolyl optionally substituted by 1 or 2 R4, quinolinyl optionally substituted by 1 or 2 R4, isoquinolinyl optionally substituted by 1 or 2 R4, quinoxalinyl optionally substituted by 1 or 2 R4, or indolyl optionally substituted by 1 or 2 R4, wherein R4 is C1-6 alkyl, aryl or heterocyclyl.
In some embodiments, Hy is pyrazine substituted by at least 1 or 2 R4, unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R4, pyridin-4-yl optionally substituted by 1 or 2 R4, pyrimidinyl optionally substituted by 1 or 2 R4, imidazolyl optionally substituted by 1 or 2 R4, thiazolyl optionally substituted by 1 or 2 R4, pyrazolyl optionally substituted by 1 or 2 R4, quinolinyl optionally substituted by 1 or 2 R4, isoquinolinyl optionally substituted by 1 or 2 R4, quinoxalinyl optionally substituted by 1 or 2 R4, or indolyl optionally substituted by 1 or 2 R4, wherein R4 is C1-6 alkyl, aryl or heterocyclyl.
In some embodiments, Hy is selected from:
Figure US08283367-20121009-C00005
Figure US08283367-20121009-C00006
In some embodiments, R1a is H.
In some embodiments, RB is H.
In some embodiments, R4 is unsubstituted.
In some embodiments, R4 is methyl, ethyl, propyl, butyl, aryl optionally substituted with 1, 2 or 3 R5, or heteroaryl optionally substituted with 1, 2 or 3 R5.
In some embodiments, R4 is C1-6 alkyl, aryl or heterocyclyl.
In some embodiments, R4 is methyl, butyl, phenyl, thienyl, or morpholino.
In some embodiments:
Z is —CH(OH)CH3; and
Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R4.
In some embodiments:
Z is —CH(OH)CH3; and
Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 R4.
In some embodiments:
Z is —CH2NHR1;
Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R4; and
R1 is carbocyclyl-C(═O)— or carbocyclyl-S(═O)2—, each optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3.
In some embodiments:
Z is —CH2NHR1;
Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 R4; and
R1 is carbocyclyl-C(═O)— or carbocyclyl-S(═O)2—, each optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, F, Cl, Br, I, C1-4 haloalkyl, carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3, and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3.
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3 or —CH2NHR1;
Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 R4;
R1 is carbocyclyl-C(═O)— or heterocyclyl-C(═O)—, each optionally substituted with 1, 2 or 3 R3;
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
R4 is, independently, selected from C1-6 alkyl, aryl and heterocyclyl;
with the proviso that when Z is —CH(OH)CH3and Q is
Figure US08283367-20121009-C00007

then Hy is other than
Figure US08283367-20121009-C00008
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3 or —CH2NHR1;
Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 C1-6 alkyl, aryl or heterocyclyl;
R1 is carbocyclyl-C(═O)— or heterocyclyl-C(═O)—, each optionally substituted with 1, 2 or 3 R3;
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
R4 is, independently, selected from C1-6 alkyl, aryl and heterocyclyl;
with the proviso that when Z is —CH(OH)CH3and Q is
Figure US08283367-20121009-C00009

then Hy is other than
Figure US08283367-20121009-C00010
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3 or —CH2NHR1;
Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 C1-6 alkyl, phenyl, thienyl or morpholino;
R1 is carbocyclyl-C(═O)— or heterocyclyl-C(═O)—, each optionally substituted with 1, 2 or 3 R3;
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
R4 is, independently, selected from C1-6 alkyl, aryl and heterocyclyl;
with the proviso that when Z is —CH(OH)CH3and Q is
Figure US08283367-20121009-C00011

then Hy is other than
Figure US08283367-20121009-C00012
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3 or —CH2NHR1;
Hy is selected from
Figure US08283367-20121009-C00013
Figure US08283367-20121009-C00014
R1 is carbocyclyl-C(═O)— or heterocyclyl-C(═O)—, each optionally substituted with 1, 2 or 3 R3; and
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—.
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3 or —CH2NHR1;
Hy is selected from
Figure US08283367-20121009-C00015
Figure US08283367-20121009-C00016
R1 is —CO-(4-methylphenyl).
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(OH)2 or pinanediol boronic ester;
Z is —CH(OH)CH3 or —CH2NHR1;
Hy is selected from
Figure US08283367-20121009-C00017
Figure US08283367-20121009-C00018
R1 is —CO-(4-methylphenyl).
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3;
Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R4;
RB is, independently, selected from C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, —OR4a, SR4a, —CN, halo, haloalkyl, —NH2, —NH(alkyl), —N(alkyl)2, —NHC(═O)O-alkyl, —NHC(═O)alkyl, —COOH, —C(═O)O-alkyl, —C(═O)alkyl, —C(O)H, —S(═O)-alkyl, —S(═O)2-alkyl, —S(═O)-aryl, —S(═O)2-aryl, carbocyclyl optionally substituted with 1, 2 or 3 R5, and heterocyclyl optionally substituted with 1, 2 or 3 R5;
R4a is H, C1-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, carbocyclyl or heterocyclyl;
R5 is, independently, selected from C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
with the proviso that when Q is
Figure US08283367-20121009-C00019

then Hy is other than
Figure US08283367-20121009-C00020
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3;
Hy is pyrazine substituted by at least 1 or 2 R4, unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R4, pyridin-4-yl optionally substituted by 1 or 2 R4, N-oxo-pyridinyl optionally substituted by 1 or 2 R4, pyrimidinyl optionally substituted by 1 or 2 R4, imidazolyl optionally substituted by 1 or 2 R4, thiazolyl optionally substituted by 1 or 2 R4, oxazolyl optionally substituted by 1 or 2 R4, pyrrolyl optionally substituted by 1 or 2 R4, pyrazolyl optionally substituted by 1 or 2 R4, quinolinyl optionally substituted by 1 or 2 R4, isoquinolinyl optionally substituted by 1 or 2 R4, quinoxalinyl optionally substituted by 1 or 2 R4, indolyl optionally substituted by 1 or 2 R4, quinazolinyl optionally substituted by 1 or 2 R4, benzoimidazolyl optionally substituted by 1 or 2 R4, benzothiazolyl optionally substituted by 1 or 2 R4, or benzoxazolyl optionally substituted by 1 or 2 R4;
R4 is, independently, selected from C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, —OR4a, —SR4a, —CN, halo, haloalkyl, —NH2, —NH(alkyl), —N(alkyl)2, —NHC(═O)O-alkyl, —NHC(═O)alkyl, —COOH, —C(═O)O-alkyl, —C(═O)alkyl, —C(O)H, —S(═O)-alkyl, —S(═O)2-alkyl, —S(═O)-aryl, —S(═O)2-aryl, carbocyclyl optionally substituted with 1, 2 or 3 R5, and heterocyclyl optionally substituted with 1, 2 or 3 R5;
R4a is H, C1-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, carbocyclyl or heterocyclyl; and
R5 is, independently, selected from C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—.
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3;
Hy is pyrazine substituted by at least 1 or 2 R4, unsubstituted pyridin-2yl, pyridin-3-yl optionally substituted by 1 or 2 R4, pyridin-4-yl optionally substituted by 1 or 2 R4, N-oxo-pyridinyl optionally substituted by 1 or 2 R4, pyrimidinyl optionally substituted by 1 or 2 R4, imidazolyl optionally substituted by 1 or 2 R4, thiazolyl optionally substituted by 1 or 2 R4, pyrazolyl optionally substituted by 1 or 2 R4, quinolinyl optionally substituted by 1 or 2 R4, isoquinolinyl optionally substituted by 1 or 2 R4, quinoxalinyl optionally substituted by 1 or 2 R4, or indolyl optionally substituted by 1 or 2 R4;
R4 is, independently, selected from C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, —OR4a, —SR4a, —CN, halo, haloalkyl, —NH2, —NH(alkyl), —N(alkyl)2, —NHC(═O)O-alkyl, —NHC(═O)alkyl, —COOH, —C(═O)O-alkyl, —C(═O)alkyl, —C(O)H, —S(═O)-alkyl, —S(═O)2-alkyl, —S(═O)-aryl, —S(═O)2-aryl, carbocyclyl optionally substituted with 1, 2 or 3 R5, and heterocyclyl optionally substituted with 1, 2 or 3 R5;
R4a is H, C1-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, carbocyclyl or heterocyclyl; and
R5 is, independently, selected from C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—.
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3; and
Hy is selected from
Figure US08283367-20121009-C00021
Figure US08283367-20121009-C00022
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(OH)2 or pinanediol boronic ester;
Z is —CH(OH)CH3; and
Hy is selected from
Figure US08283367-20121009-C00023
Figure US08283367-20121009-C00024
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH2NR1aR1;
Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R4;
R1 is H, C1-10 alkyl, carbocyclyl, heterocyclyl, C1-10 alkyl-C(═O)—, C2-10 alkenyl-C(═O)—, C2-10 alkynyl-C(═O)—, carbocyclyl-C(═O)—, heterocyclyl-C(═O)—, carbocyclylalkyl-C(═O)—, heterocyclylalkyl-C(═O)—, C1-10 alkyl-S(═O)2—, carbocyclyl-S(═O)2—, heterocyclyl-S(═O)2—, carbocyclylalkyl-S(═O)2—, heterocyclylalkyl-S(═O)2—, C1-C10 alkyl-NHC(═O)—, carbocyclyl-NHC(═O)—, heterocyclyl-NHC(═O)—, carbocyclylalkyl-NHC(═O)—, heterocyclylalkyl-NHC(═O)—, C1-C10 alkyl-OC(═O)—, carbocyclyl-OC(═O)—, heterocyclyl-OC(═O)—, carbocyclylalkyl-OC(═O)—, heterocyclylalkyl-OC(═O)—, C1-10 alkyl-NH—C(═O)—NHS(═O)2—, carbocyclyl-NH—C(═O)—NHS(═O)2—, heterocyclyl-NH—C(═O)—NHS(═O)2—, C1-10 alkyl-S(═O)2—NH—C(═O)—, carbocyclyl-S(═O)2—NH—C(═O)—, heterocyclyl-S(═O)2—NH—C(═O)—, or an amino protecting group; wherein R1 is optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3;
R1a is H. Alternatively, R1a and R1 together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocyclyl group optionally substituted with 1, 2, or 3 R3;
R2 is, independently, H or C1-6 alkyl;
alternatively, two R2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group;
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
R4 is, independently, selected from C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, —OR4a, —SR4a, —CN, halo, haloalkyl, —NH2, —NH(alkyl), —N(alkyl)2, —NHC(═O)O-alkyl, —NHC(═O)alkyl, —COOH, —C(═O)O-alkyl, —C(═O)alkyl, —C(O)H, —S(═O)-alkyl, —S(═O)2-alkyl, —S(═O)-aryl, —S(═O)2-aryl, carbocyclyl optionally substituted with 1, 2 or 3 R5, and heterocyclyl optionally substituted with 1, 2 or 3 R5;
R4a is H, C1-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, carbocyclyl or heterocyclyl; and
R5 is, independently, selected from C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—.
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH2NR1aR1;
Hy is pyrazine substituted by at least 1 or 2 R4, pyridinyl optionally substituted by 1 or 2 R4, N-oxo-pyridinyl optionally substituted by 1 or 2 R4, pyrimidinyl optionally substituted by 1 or 2 R4, imidazolyl optionally substituted by 1 or 2 R4, thiazolyl optionally substituted by 1 or 2 R4, oxazolyl optionally substituted by 1 or 2 R4, pyrrolyl optionally substituted by 1 or 2 R4, pyrazolyl optionally substituted by 1 or 2 R4, quinolinyl optionally substituted by 1 or 2 R4, isoquinolinyl optionally substituted by 1 or 2 R4, quinoxalinyl optionally substituted by 1 or 2 R4, indolyl optionally substituted by 1 or 2 R4, quinazolinyl optionally substituted by 1 or 2 R4, benzoimidazolyl optionally substituted by 1 or 2 R4, benzothiazolyl optionally substituted by 1 or 2 R4, or benzoxazolyl optionally substituted by 1 or 2 R4;
R1 is H, C1-10 alkyl, carbocyclyl, heterocyclyl, C1-10 alkyl-C(═O)—, C2-10 alkenyl-C(═O)—, C2-10 alkynyl-C(═O)—, carbocyclyl-C(═O)—, heterocyclyl-C(═O)—, carbocyclylalkyl-C(═O)—, heterocyclylalkyl-C(═O)—, C1-10 alkyl-S(═O)2—, carbocyclyl-S(═O)2—, heterocyclyl-S(═O)2—, carbocyclylalkyl-S(═O)2—, heterocyclylalkyl-S(═O)2—, C1-C10 alkyl-NHC(═O)—, carbocyclyl-NHC(═O)—, heterocyclyl-NHC(═O)—, carbocyclylalkyl-NHC(═O)—, heterocyclylalkyl-NHC(═O)—, C1-C10 alkyl-OC(═O)—, carbocyclyl-OC(═O)—, heterocyclyl-OC(═O)—, carbocyclylalkyl-OC(═O)—, heterocyclylalkyl-OC(═O)—, C1-10 alkyl-NH—C(═O)—NHS(═O)2—, carbocyclyl-NH—C(═O)—NHS(═O)2—, heterocyclyl-NH—C(═O)—NHS(═O)2—, C1-10 alkyl-S(═O)2—NH—C(═O)—, carbocyclyl-S(═O)2—NH—C(═O)—, heterocyclyl-S(═O)2—NH—C(═O)—, or an amino protecting group; wherein R1 is optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3;
R1a is H. Alternatively, R1a and R1 together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocyclyl group optionally substituted with 1, 2, or 3 R3;
R2 is, independently, H or C1-6 alkyl;
alternatively, two R2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group;
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
R4 is, independently, selected from C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, —OR4a, SR4a, —CN, halo, haloalkyl, —NH2, —NH(alkyl), —N(alkyl)2, —NHC(═O)O-alkyl, —NHC(═O)alkyl, —COOH, —C(═O)O-alkyl, —C(═O)alkyl, —C(O)H, —S(═O)-alkyl, —S(═O)2-alkyl, —S(═O)-aryl, —S(═O)2-aryl, carbocyclyl optionally substituted with 1, 2 or 3 R5, and heterocyclyl optionally substituted with 1, 2 or 3 R5;
R4a is H, C1-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, carbocyclyl or heterocyclyl; and
R5 is, independently, selected from C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—.
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH2NR1aR1;
Hy is selected from
Figure US08283367-20121009-C00025
Figure US08283367-20121009-C00026
R1 is H, C1-10 alkyl, carbocyclyl, heterocyclyl, C1-10 alkyl-C(═O)—, C2-10 alkenyl-C(═O)—, C2-10 alkynyl-C(═O)—, carbocyclyl-C(═O)—, heterocyclyl-C(═O)—, carbocyclylalkyl-C(═O)—, heterocyclylalkyl-C(═O)—, C1-10 alkyl-S(═O)2—, carbocyclyl-S(═O)2—, heterocyclyl-S(═O)2—, carbocyclylalkyl-S(═O)2—, heterocyclylalkyl-S(═O)2—, C1-C10 alkyl-NHC(═O)—, carbocyclyl-NHC(═O)—, heterocyclyl-NHC(═O)—, carbocyclylalkyl-NHC(═O)—, heterocyclylalkyl-NHC(═O)—, C1-C10 alkyl-OC(═O)—, carbocyclyl-OC(═O)—, heterocyclyl-OC(═O)—, carbocyclylalkyl-OC(═O)—, heterocyclylalkyl-OC(═O)—, C1-10 alkyl-NH—C(═O)—NHS(═O)2—, carbocyclyl-NH—C(═O)—NHS(═O)2—, heterocyclyl-NH—C(═O)—NHS(═O)2—, C1-10 alkyl-S(═O)2—NH—C(═O)—, carbocyclyl-S(═O)2—NH—C(═O)—, heterocyclyl-S(═O)2—NH—C(═O)—, or an amino protecting group; wherein R1 is optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3;
R1a is H. Alternatively, R1a and R1 together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocyclyl group optionally substituted with 1, 2, or 3 R3;
R2 is, independently, H or C1-6 alkyl;
alternatively, two R2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group; and
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—.
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH2NHR1;
Hy is selected from
Figure US08283367-20121009-C00027
Figure US08283367-20121009-C00028
R1 is H, C1-10 alkyl, carbocyclyl, heterocyclyl, C1-10 alkyl-C(═O)—, C2-10 alkenyl-C(═O)—, C2-10 alkynyl-C(═O)—, carbocyclyl-C(═O)—, heterocyclyl-C(═O)—, carbocyclylalkyl-C(═O)—, heterocyclylalkyl-C(═O)—, C1-10 alkyl-S(═O)2—, carbocyclyl-S(═O)2—, heterocyclyl-S(═O)2—, carbocyclylalkyl-S(═O)2—, heterocyclylalkyl-S(═O)2—, C1-C10 alkyl-NHC(═O)—, carbocyclyl-NHC(═O)—, heterocyclyl-NHC(═O)—, carbocyclylalkyl-NHC(═O)—, heterocyclylalkyl-NHC(═O)—, C1-C10 alkyl-OC(═O)—, carbocyclyl-OC(═O)—, heterocyclyl-OC(═O)—, carbocyclylalkyl-OC(═O)—, heterocyclylalkyl-OC(═O)—, C1-10 alkyl-NH—C(═O)—NHS(═O)2—, carbocyclyl-NH—C(═O)—NHS(═O)2—, heterocyclyl-NH—C(═O)—NHS(═O)2—, C1-10 alkyl-S(═O)2—NH—C(═O)—, carbocyclyl-S(═O)2—NH—C(═O)—, heterocyclyl-S(═O)2—NH—C(═O)—, or an amino protecting group; wherein R1 is optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3;
R2 is, independently, H or C1-6 alkyl;
alternatively, two R2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group; and
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—.
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH2NHR1;
Hy is selected from
Figure US08283367-20121009-C00029
Figure US08283367-20121009-C00030
R1 is carbocyclyl-C(═O)—, heterocyclyl-C(═O)—, wherein R1 is optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3;
R2 is, independently, H or C1-6 alkyl;
alternatively, two R2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group; and
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—.
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH2NHR1;
Hy is selected from
Figure US08283367-20121009-C00031
Figure US08283367-20121009-C00032
R1 is aryl-C(═O)—, wherein R1 is optionally substituted with 1 or 2 substituents selected from C1-6 alkyl, F, Cl, Br, I, and C1-4 haloalkyl.
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH2NHR1;
Hy is selected from
Figure US08283367-20121009-C00033
Figure US08283367-20121009-C00034
R1 is —CO-(4-methylphenyl).
In some embodiments, compounds of the invention include compounds of Formula (I) or pharmaceutically acceptable salt forms thereof, wherein:
Q is —B(OH)2 pinanediol boronic ester;
Z is —CH2NHR1;
Hy is selected from
Figure US08283367-20121009-C00035
Figure US08283367-20121009-C00036
R1 is —CO-(4-methylphenyl).
In some embodiments compounds of the invention include compounds of Formula (I):
Figure US08283367-20121009-C00037

or pharmaceutically acceptable salt forms thereof, wherein:
  • Q is —B(ORB)2, boronic acid, or a cyclic boronic ester wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3 or —CH2NR1aR1;
Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R4;
R1 is H, C1-10 alkyl, carbocyclyl, heterocyclyl, C1-10 alkyl-C(═O)—, C2-10 alkenyl-C(═O)—, C2-10 alkynyl-C(═O)—, carbocyclyl-C(═O)—, heterocyclyl-C(═O)—, carbocyclylalkyl-C(═O)—, heterocyclylalkyl-C(═O)—, C1-10 alkyl-S(═O)2—, carbocyclyl-S(═O)2—, heterocyclyl-S(═O)2—, carbocyclylalkyl-S(═O)2—, heterocyclylalkyl-S(═O)2—, C1-C10 alkyl-NHC(═O)—, carbocyclyl-NHC(═O)—, heterocyclyl-NHC(═O)—, carbocyclylalkyl-NHC(═O)—, heterocyclylalkyl-NHC(═O)—, C1-C10 alkyl-OC(═O)—, carbocyclyl-OC(═O)—, heterocyclyl-OC(═O)—, carbocyclylalkyl-OC(═O)—, heterocyclylalkyl-OC(═O)—, C1-10 alkyl-NH—C(═O)—NHS(═O)2—, carbocyclyl-NH—C(═O)—NHS(═O)2—, heterocyclyl-NH—C(═O)—NHS(═O)2—, C1-10 alkyl-S(═O)2—NH—C(═O)—, carbocyclyl-S(═O)2—NH—C(═O)—, heterocyclyl-S(═O)2—NH—C(═O)—, or an amino protecting group; wherein R1 is optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3;
R1a is H. Alternatively, R1a and R1 together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocyclyl group optionally substituted with 1, 2, or 3 R3;
R2 is, independently, H or C1-6 alkyl;
alternatively, two R2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group;
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, (alkyl-O)r-alkyl, HO-(alkyl-O)r-alkyl-, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
R4 is, independently, selected from C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, —OR4a, —SR4a, —CN, halo, haloalkyl, —NH2, —NH(alkyl), —N(alkyl)2, —NHC(═O)O-alkyl, —NHC(═O)alkyl, —COOH, —C(═O)O-alkyl, —C(═O)alkyl, —C(O)H, —S(═O)-alkyl, —S(═O)2-alkyl, —S(═O)-aryl, —S(═O)2-aryl, carbocyclyl optionally substituted with 1, 2 or 3 R5, and heterocyclyl optionally substituted with 1, 2 or 3 R5;
R4a is H, C1-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, carbocyclyl or heterocyclyl;
R5 is, independently, selected from C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, (alkyl-O)r-alkyl, HO-(alkyl-O)r-alkyl-, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
r is 0, 1, 2, 3 or 4;
with the proviso that when Z is —CH(OH)CH3and Q is
Figure US08283367-20121009-C00038

then Hy is other than
Figure US08283367-20121009-C00039
It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.
As used herein, the phrase “boronic acid” refers to a compound containing a B(OH)2 moiety. In some embodiments, boronic acid compounds can form oligomeric anhydrides by dehydration of the boronic moiety. For example, Snyder, et al., J. Am. Chem. Soc., 1958, 80, 3611 report oligomeric arylboronic acids. Thus, unless otherwise indicated, “boronic acid”, or a chemical formula containing a —B(OH)2 moiety, is intended to encompass free boronic acids, oligomeric anhydrides, including but not limited to, dimers, trimers, tetramers, and mixtures thereof.
As used herein, “boronic acid anhydride” or “boronic anhydride” refers to a compound formed by the combination of two or more molecules of a boronic acid compound of Formula (I), with loss of one or more water molecules from the boronic acid moieties. When contacted with water, the boronic acid anhydride compound can be hydrated to release free boronic acid compound. In some embodiments, the boronic acid anhydride structure can contain two, three, four, or more boronic acid units and can have a cyclic or linear configuration. In some embodiments, the boronic acid anhydride compound exists substantially in a single oligomeric form; however, boronic acid anhydrides also encompass mixtures of different oligomeric boronic acid anhydride as well as free boronic acids.
Non-limiting examples of boronic acid anhydrides of the invention include compounds of Formula (II) and (III) where G is a moiety of Formula (IV) and t is 0 to 10 or 1, 2, 3, or 4.
Figure US08283367-20121009-C00040
In some embodiments, at least about 80% of boronic acid present in a boronic acid anhydride compound exists in a single oligomeric anhydride form. In further embodiments, at least about 85, about 90, about 95, or about 99% of the boronic acid present in the boronic acid anhydride exists in a single oligomeric anhydride form. In some embodiments, the boronic acid anhydride compound consists essentially of a single oligomeric boronic acid anhydride. In yet further embodiments, the boronic acid anhydride compound consists of a single oligomeric boronic acid anhydride. In further embodiments, the boronic acid anhydride compound contains a boroxine of Formula (III), wherein t is 1.
Boronic acid anhydride compounds can be prepared from the corresponding boronic acid compound by exposure to dehydrating conditions, including, for example, crystallization, lyophilization, exposure to heat, and/or exposure to a drying agent. Some suitable crystallization solvents include ethyl acetate, dichloromethane, hexanes, ether, benzene, acetonitrile, ethanol, and mixtures thereof.
As used herein, the phrase “boronic ester” or “boronic acid ester” refers to an ester derivative of a boronic acid compound. As used herein, “cyclic boronic ester” is intended to mean a stable cyclic boronic moiety of general formula —B(OR)(OR) wherein the two R substituents are linked together forming a cyclic moiety (e.g., 3- to 10-membered cycloalkyl group) optionally further substituted with one or more substituents or fused with (sharing at least one bond) one or more further carbocyclyl or heterocarbocyclyl groups. The cyclic boronic ester can contain from 2 to 20 carbon atoms, and optionally, a heteroatom which can be N, S, or O. Cyclic boronic esters are well known in the art. Examples of cyclic boronic esters include, but are not limited to, pinanediol boronic ester, pinacol boronic ester, 1,2-ethanediol boronic ester, 1,3-propanediol boronic ester, 1,2-propanediol boronic ester, 2,3-butanediol boronic ester, 1,1,2,2-tetramethylethanediol boronic ester, 1,2-diisopropylethanediol boronic ester, 5,6-decanediol boronic ester, 1,2-dicyclohexylethanediol boronic ester, bicyclohexyl-1,1′-diol, diethanolamine boronic ester, and 1,2-diphenyl-1,2-ethanediol boronic ester.
In some embodiments, the “cyclic boronic ester” has Formula (II-a):
Figure US08283367-20121009-C00041

wherein:
D is absent, O, S, NR16, or CR15eR15f,
R15a, R15b, R15c, R15d, R15e, R15f are each, independently, H, C1-C10 alkyl C3-C7 cycloalkyl, aryl or heteroaryl, wherein said C1-C10 alkyl, C3-C10 cycloalkyl, aryl or heteroaryl are each optionally substituted by 1, 2, 3 or 4 halo, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl;
or R15a and R15b together with the C atoms to which they are attached form C3-C10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl;
or R15c and R15d together with the C atoms to which they are attached form C3-C10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl;
or R15b and R15c together with the C atoms to which they are attached and the intevening D moiety form aryl, heteroaryl, C3-C10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl;
R16 is H or C1-C6 alkyl; and
p and q are each, independently, 1, 2 or 3.
In some embodiments, D is absent.
In some embodiments, D is NR16.
In some embodiments, D is NH.
In some embodiments, D is CH2.
In some embodiments, R15a and R15b together with the C atoms to which they are attached form C3-C10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl; and R15c and R15d together with the C atoms to which they are attached form C3-C10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl.
In some embodiments, R15a and R15b together with the C atoms to which they are attached form cyclopropyl, cyclobutyl, cyclopenytyl, cyclohexyl or cycloheptyl; and R15c and R15d together with the C atoms to which they are attached form cyclopropyl, cyclobutyl, cyclopenytyl, cyclohexyl or cycloheptyl.
In some embodiments, D is absent and R15b and R15c together with the C atoms to which they are attached form aryl, heteroaryl, C3-C10 cycloalkyl or a 3- to 10-membered heterocycloalkyl group, each optionally substituted by 1, 2, 3 or 4 halo, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl.
In some embodiments, D is absent and R15b and R15c together with the C atoms to which they are attached form C3-C10 cycloalkyl optionally substituted by 1, 2, 3 or 4 halo, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, OH, amino, alkylamino, dialkylamino, aryl, or heteroaryl.
In some embodiments, D is absent and R15b and R15c together with the C atoms to which they are attached form C3-C10 cycloalkyl optionally substituted by 1, 2, 3 or 4 halo or C1-C4 alkyl.
In some embodiments, D is absent and R15b and R15c together with the C atoms to which they are attached form a C7-C10 bicyclic cycloalkyl group optionally substituted by 1, 2, 3 or 4 halo or C1-C4 alkyl.
In some embodiments, p and q are each 1.
In some embodiments, at least one of R15a, R15b, R15c, R15d is other than H.
Further examples of “cyclic boronic esters”, as defined herein, include, boronic esters with the following structures:
Figure US08283367-20121009-C00042

wherein: W is a substituted or unsubstituted C4-C10 cycloalkyl ring or a substituted or unsubstituted phenyl ring; W1 is, independently at each occurrence, a substituted or unsubstituted C3-C6 cycloalkyl ring. Groups R15a, R15b, R15c, R15d, R15e, R15f, p and q are, defined as provided above.
As used herein, the term “alkyl” or “alkylene” is meant to refer to a saturated hydrocarbon group which is straight-chained or branched. Example alkyl groups include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, s-butyl, t-butyl), pentyl (e.g., n-pentyl, isopentyl, neopentyl) and the like. An alkyl group can contain from 1 to about 20, from 2 to about 20, from 1 to about 10, from 1 to about 8, from 1 to about 6, from 1 to about 4, or from 1 to about 3 carbon atoms.
As used herein, “alkenyl” refers to an alkyl group having one or more double carbon-carbon bonds. Example alkenyl groups include ethenyl, propenyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl, and the like.
As used herein, “alkynyl” refers to an alkyl group having one or more triple carbon-carbon bonds. Example alkynyl groups include ethynyl, propynyl, butynyl, pentynyl, and the like.
As used herein, “haloalkyl” refers to an alkyl group having one or more halogen substituents. Example haloalkyl groups include CF3, C2F5, CHF2, CCl3, CHCl2, C2Cl5, and the like. An alkyl group in which all of the hydrogen atoms are replaced with halogen atoms can be referred to as “perhaloalkyl.” Examples perhaloalkyl groups include CF3and C2F5.
As used herein, “carbocyclyl” groups are saturated (i.e., containing no double or triple bonds) or unsaturated (i.e., containing one or more double or triple bonds) cyclic hydrocarbon moieties. Carbocyclyl groups can be mono- or polycyclic. Example carbocyclyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, 1,3-cyclopentadienyl, cyclohexenyl, norbornyl, norpinyl, norcarnyl, adamantyl, phenyl, and the like. Carbocyclyl groups can be aromatic (e.g., “aryl”) or non-aromatic (e.g., “cycloalkyl”). In some embodiments, carbocyclyl groups can have from 3 to about 20, 3 to about 10, or 3 to about 7 carbon atoms.
As used herein, “aryl” refers to aromatic carbocyclyl groups including monocyclic or polycyclic aromatic hydrocarbons such as, for example, phenyl, naphthyl, anthracenyl, phenanthrenyl, indanyl, indenyl, and the like. In some embodiments, aryl groups have from 6 to about 18 ring-forming carbon atoms.
As used herein, “cycloalkyl” refers to non-aromatic carbocyclyl groups including cyclized alkyl, alkenyl, and alkynyl groups. Cycloalkyl groups can include bi- or poly-cyclic ring systems. Example cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptatrienyl, norbornyl, norpinyl, norcarnyl, adamantyl, and the like. Also included in the definition of cycloalkyl are moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the cycloalkyl ring, for example, benzo derivatives of cyclopentane (indanyl), cyclohexane (tetrahydronaphthyl), and the like. Also included in the definition of cycloalkyl are groups in which one or more of the ring-forming carbon atoms is substituted by an oxo group. In some embodiments, cycloalkyl groups can have 3, 4, 5, 6, or 7 ring forming carbon atoms. In some embodiments, cycloalkyl groups can have 0, 1, or 2 double or triple ring-forming bonds.
As used herein, “heterocyclyl” groups can be saturated or unsaturated carbocyclyl groups wherein one or more of the ring-forming carbon atoms of the carbocyclyl group is replaced with a heteroatom such as O, S, or N. Heterocyclyl groups can be aromatic (e.g., “heteroaryl”) or non-aromatic (e.g., “heterocycloalkyl”). Heterocyclyl groups can correspond to hydrogenated and partially hydrogenated heteroaryl groups. Heterocyclyl groups can contain, in addition to at least one heteroatom, from about 1 to about 20, about 2 to about 10, or about 2 to about 7 carbon atoms and can be attached through a carbon atom or heteroatom. Additionally, any ring-forming carbon atom or heteroatom can be substituted by one or two oxo or sulfide groups. Examples of heterocyclyl groups include morpholino, thiomorpholino, piperazinyl, tetrahydrofuranyl, tetrahydrothienyl, 2,3-dihydrobenzofuryl, 1,3-benzodioxole, benzo-1,4-dioxane, piperidinyl, pyrrolidinyl, isoxazolidinyl, isothiazolidinyl, pyrazolidinyl, oxazolidinyl, thiazolidinyl, imidazolidinyl, and the like. In some embodiments, the heterocyclyl group is a 5- or 6-membered heterocyclyl group.
As used herein, “heteroaryl” groups are aromatic heterocarbocyclyl groups and include monocyclic and polycyclic aromatic hydrocarbons that have at least one heteroatom ring member such as sulfur, oxygen, or nitrogen. Heteroaryl groups include, without limitation, pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrrolyl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl indazolyl, 1,2,4-thiadiazolyl, isothiazolyl, benzothienyl, purinyl, carbazolyl, benzimidazolyl, and the like. In some embodiments, heteroaryl groups can have from 3 to about 20 ring-forming carbon atoms, and in further embodiments from about 3 to about 12 ring forming carbon atoms. In some embodiments, heteroaryl groups have 1 to about 4, 1 to about 3, or 1 to 2 heteroatoms. In some embodiments, the heteroaryl group has at least one ring-forming N atom.
As used herein, “heterocycloalkyl” refers to a non-aromatic heterocyclyl group including cyclized alkyl, alkenyl, and alkynyl groups where one or more of the ring-forming carbon atoms is replaced by a heteroatom such as an O, N, or S atom. Ring-forming carbon and heteroatoms such as S and N can further be oxidized in a heterocycloalkyl moeity. For example, the ring-forming carbon or heteroatom can bear one or two oxo or sufido moieties (e.g., >C═O, >S═O, >S(═O)2, N→O, etc.). Also included in the definition of heterocycloalkyl are moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the nonaromatic heterocyclic ring, for example phthalimidyl, naphthalimidyl pyromellitic diimidyl, phthalanyl, and benzo derivatives of saturated heterocycles such as indolene and isoindolene groups. In some embodiments, heterocycloalkyl groups have 3 to about 20 ring-forming atoms. In some embodiments, heterocycloalkyl groups have 3, 4, 5, 6, or 7 ring-forming atoms. In some embodiments, heterocycloalkyl groups have 0, 1, or 2 double or triple ring-forming bonds.
As used herein, “halo” or “halogen” includes fluoro, chloro, bromo, and iodo.
As used herein, “alkoxy” refers to an —O-alkyl group. Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the like. In some embodiments, alkoxy groups have from 1 to 20, 1 to 12, 1 to 8, 1 to 6, 1 to 4 or 1 to 3 carbon atoms.
As used herein, “alkoxyalkoxy” refers to an —O-alkyl-O-alkyl group.
As used herein, “thioalkoxy” refers to an alkoxy group in which the O atom is replaced by an S atom.
As used herein, “aryloxy” refers to an —O-aryl group. An example aryloxy group is phenoxy.
As used herein, “thioaryloxy” refers to an aryloxy group in which the O atom is replaced by an S atom.
As used herein, “aralkyl” refers to an alkyl moiety substituted by an aryl group. Example aralkyl groups include benzyl and naphthylmethyl groups. In some embodiments, aralkyl groups have from 7 to 11 carbon atoms.
As used herein, “amino” refers to an —NH2 group. “Alkylamino” refers to an amino group substituted by an alkyl group and “dialkylamino” refers to an amino group substituted by two alkyl groups. On the contrary, “aminoalkyl” refers to an alkyl group substituted by an amino group.
As used herein, “carbonyl” refers to >C═O.
As used herein, “carboxy” or “carboxyl” refers to —COOH.
As used herein, “hydroxy” refers to —OH.
As used herein, “mercapto” refers to —SH.
As used herein, “ureido” refers to —NHCONH2.
As used herein, “sulfinyl” refers to >SO.
As used herein, “sulfonyl” refers to >SO2.
As used herein, “oxy” refers to —O—.
The above chemical terms can be combined to refer to moieties containing a combination of chemical groups. This combination term is generally read such that a recited term is understood to be a substituent of a following term. For example, “alkylcarbonylalkenyl” refers to an alkenyl group substituted by a carbonyl group which in turn is substituted by an alkyl group. The following terms can also exemplify such combinations.
As used herein, “carbocyclylalkyl” refers to an alkyl moiety substituted by a carbocyclyl group. Example carbocyclylalkyl groups include “aralkyl” (alkyl substituted by aryl) and “cycloalkylalkyl” (alkyl substituted by cycloalkyl).
As used herein, “carbocyclylalkenyl” refers to an alkenyl moiety substituted by a carbocyclyl group. Example carbocyclylalkenyl groups include “aralkenyl” (alkenyl substituted by aryl) and “cycloalkylalkenyl” (alkenyl substituted by cycloalkyl).
As used herein, “carbocyclylalkynyl” refers to an alkynyl moiety substituted by a carbocyclyl group. Example carbocyclylalkynyl groups include “aralkynyl” (alkynyl substituted by aryl) and “cycloalkylalkynyl” (alkynyl substituted by cycloalkyl).
As used herein, “heterocyclylalkyl” refers to an alkyl moiety substituted by a heterocarbocyclyl group. Example heterocyclylalkyl groups include “heteroarylalkyl” (alkyl substituted by heteroaryl) and “heterocycloalkylalkyl” (alkyl substituted by heterocycloalkyl).
As used herein, “heterocyclylalkenyl” refers to an alkenyl moiety substituted by a heterocyclyl group. Example heterocarbocyclylalkenyl groups include “heteroarylalkenyl”(alkenyl substituted by heteroaryl) and “heterocycloalkylalkenyl” (alkenyl substituted by heterocycloalkyl).
As used herein, “heterocyclylalkynyl” refers to an alkynyl moiety substituted by a heterocarbocyclyl group. Example heterocyclylalkynyl groups include “heteroarylalkynyl”(alkynyl substituted by heteroaryl) and “heterocycloalkynylalkyl” (alkynyl substituted by heterocycloalkyl).
As used herein, the phrase “5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom” refers to 5- or 6-membered heterocycloalkyl groups or 5- or 6-membered heteroaryl groups, each containing within the 5- or 6-membered ring at least one N atom and optionally additional heteroatoms (e.g., 1, 2 or 3additional heteroatoms selected from N, O and S). Example 6-membered heterocyclic groups containing at least one ring-forming N atom include aromatics such as pyridyl (i.e., pryidinyl), pyrimidinyl, pyrazinyl, triazinyl, and the like. Example 6-membered heterocyclic groups containing at least one ring-forming N atom include non-aromatics such as piperidinyl, piperazinyl, morpholino, and the like. Example 5-membered heterocyclic groups containing at least one ring-forming N atom include aromatics such as imidazolyl, pyrrolyl, pyrrazolyl, trizaolyl, oxoazoyl, thiazolyl and the like. Example 5-membered heterocyclic groups containing at least one ring-forming N atom include non-aromatics such as pyrrolidine, imidazolino, imidazolidino, and the like. The 5- or 6-membered ring can be attached through either a C atom or a heteroatom. As stated above, the 5- or 6-membered ring can be fused to an aryl or heteroaryl group, meaning that the 5- or 6-membered ring shares a bond with an aryl or heteroaryl group. The fused aryl or heteroaryl group can be a 5- or 6-membered group such as phenyl, naphthyl, pyridyl, pyimidinyl, pyrazinyl, triazinyl, imidazolyl, pyrrolyl, pyrrazolyl, trizaolyl, oxoazoyl, thiazolyl and the like. Example 5- or 6-membered heterocyclic groups containing at least one ring-forming N atom and which are fused to an aryl or heteroaryl group include quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, benzoxazolyl and the like. The 5- or 6-membered heterocyclic group containing at least one ring-forming N atom and which are fused to an aryl or heteroaryl are preferably attached through a C or heteroatom of the 5- or 6-membered heterocyclic group.
As used herein, the phrase “protecting group” refers to a chemical functional group that can be selectively appended to and removed from functionalities, such as hydroxyl groups, amino groups, and carboxyl groups. Protecting groups are usually introduced into a chemical compound to render such functionality inert to chemical reaction conditions to which the compound is exposed. Any of a variety of protecting groups can be employed with the present invention. A protecting group of an amino moiety can be referred to as an “amino protecting group.” Amino protecting groups can have the formulas aryl-SO2—, alkyl-SO2—, aryl-C(═O)—, aralkyl-C(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, aralkyl-OC(═O)—, alkyl-OC(═O)—, aryl-NHC(═O)—, alkyl-NHC(═O)—, and the like, wherein said alkyl, aryl and aralkyl groups may be substituted or unsubstituted. Example amino and guanidino protecting groups can also include t-butyloxycarbonyl (BOC), fluorenylmethoxycarbonyl (Fmoc), benzyloxycarbonyl (Cbz), and a phthalimido group. Further representative protecting groups can be found in T. W. Green and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd. Ed., Wiley & Sons, Inc., New York (1999), which is incorporated herein by reference in its entirety.
As used herein, “substituted” indicates that at least one hydrogen atom of a chemical group is replaced by a non-hydrogen moiety. Example substituents include F, Cl, Br, I, C1-C6 alkyl, C1-C6 alkenyl, C1-C6, alkynyl, haloalkyl, NRERF, N3, NO2, CN, CNO, CNS, C(═O)ORE, RECO, REC(═O)O, RECONRE, RERFNCO, ureido, ORE, SRE, SO2-alkyl, SO2-aryl, and SO2—NRERF, wherein RE and RF are each, independently, H or C1-C6 alkyl. Alternatively, RE and RF may be combined, with the nitrogen to which they are attached, to form a 5 to 7 membered heterocyclic ring, for example pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, and N-methylpiperazinyl. When a chemical group herein is “substituted” it may have up to the full valance of substitution, provided the resulting compound is a stable compound or stable structure; for example, a methyl group may be substituted by 1, 2, or 3 substituents, a methylene group may be substituted by 1 or 2 substituents, a phenyl group may be substituted by 1, 2, 3, 4, or 5 substituents, and the like.
As used herein, “leaving group” refers to any group that can be replaced by a nucleophile upon nucleophilic substitution. Example leaving groups include, halo (F, Cl, Br, I), hydroxyl, alkoxy, mercapto, thioalkoxy, triflate, alkylsulfonyl, substituted alkylsulfonate, arylsulfonate, substituted arylsulfonate, heterocyclosulfonate or trichloroacetimidate. Representative examples include p-(2,4-dinitroanilino)benzenesulfonate, benzenesulfonate, methylsulfonate, p-methylbenzenesulfonate, p-bromobenzenesulfonate, trichloroacetimidate, acyloxy, 2,2,2-trifluoroethanesulfonate, imidazolesulfonyl and 2,4,6-trichlorophenyl.
As used herein “stable compound” or “stable structure” refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and preferably capable of formulation into an efficacious therapeutic agent. The present invention is directed only to stable compounds.
The compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present invention that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C═N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. Cis and trans geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms.
In addition to the above, the compounds herein described may have asymmetric centers which result in one enantiomer of a compound of Formula (I) demonstrating superior biological activity over the opposite enantiomer. When required, separation of the racemic material can be achieved by methods known in the art.
Compounds of the invention can also include tautomeric forms, such as keto-enol tautomers. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium.
The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The present invention also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, “pharmaceutically acceptable salts” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and in the Journal of Pharmaceutical Science, 66, 2 (1977), the disclosures of each of which are hereby incorporated by reference.
Synthesis
Compounds of the invention, including salts and solvates thereof, can be prepared using known organic synthesis techniques and can be synthesized according to any of numerous possible synthetic routes.
The reactions for preparing compounds of the invention can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected.
Preparation of compounds of the invention can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd. Ed., Wiley & Sons, Inc., New York (1999), which is incorporated herein by reference in its entirety.
Reactions can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
Compounds of the invention can be prepared according to methods for preparing aminoboronic acids, esters thereof, and related compounds described in the art, such as in U.S. Pat. Nos. 4,537,773, and in 5,614,649, each of which is incorporated herein by reference in its entirety. In some embodiments, the present compounds can be prepared by the sequential coupling of three fragment components (F1, F2, and F3) as described below.
F1 Fragment
Synthesis of compounds of the invention can involve a boron-containing fragment (F1) having a general structure indicated by Formula (A).
Figure US08283367-20121009-C00043

The boronic ester moiety of F1 can include, for example, a diol ester such as is indicated by the loop connecting oxygen atoms in Formula (A).
Stereochemistry at the carbon atom alpha to the boron atom in Formula (A) can be controlled using an asymmetric boronic ester group in the preparation of F1. For example, pinanediol esters of boronic acid can facilitate the preparation or stereochemically pure, or substantially stereochemically pure, F1 fragment. As an example, the F1 fragment can be prepared by reacting a compound of Formula (B) (showing a pinanediol boronic ester obtained from (+)-pinanediol) with a strong base (e.g., lithium diisopropylamide or lithium dicyclohexylamide) in the presence of dichloromethane or dibromomethane, followed by addition of a Lewis acid, (e.g., ZnCl2, ZnBr2, or FeCl3) to yield a compound of Formula (C) (where L is halo) having a newly introduced stereocenter at the carbon alpha to the boron.
Figure US08283367-20121009-C00044
The compound of Formula (C) can, in turn, be reacted with an alkali amide (e.g., lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide, and potassium bis(trimethylsilyl)amide) or other nucleophile that effectively inverts the newly formed stereocenter (such as by an SN2 type mechanism) and introduces an amine group (NR2) in place of the leaving group L (e.g., chloro), forming a compound of Formula (D) (where each R can independently be, e.g., alkyl, Si(alkyl)3, aryl, or aralkyl).
Figure US08283367-20121009-C00045
The compound of Formula (D) can be further reacted with an agent capable of converting the NR2 group to NH2, or salt thereof, to form an F1 fragment substantially capable of coupling with a further fragment through the amine. A suitable agent for converting the NR2 group to NH2 can be a protic acid such as HCl such as when R is a silyl group (e.g., trimethylsilyl).
The compound of Formula (B) can also be prepared according to a two step procedure involving reaction of a trialkoxyborane, such as triisopropoxyborane, with (1S, 2S, 3R, 5S)-(+) pinanediol, to give a mono-alkoxy [(1S, 2S, 3R, 5S)-(+) pinanediol]borane intermediate wherein two of the alkoxy groups of the trialkoxy borane are replaced by (1S, 2S, 3R, 5S)-(+) pinanediol. This mixed pinanediol alkoxy borane, upon reaction with the appropriate organometallic derivative, e.g. the Grignard reagent R′CH2MgBr (where R′ is prop-2-yl) or the alkyl lithium R′CH2Li (where R′ is prop-2-yl), gives compound (B) in good yields and purities. The process starting from triisopropoxyborane to give the intermediate mixed pinanediol isopropoxy borane (F) and the compounds of formula (B) is depicted in the following scheme:
Figure US08283367-20121009-C00046

F2 Fragment
The mid-section of compounds of the present invention can be represented by fragment F2 which couples to fragment F1 by peptide bond formation for form an F2-F1 intermediate. Methods for coupling compounds through peptide bonds, or amide bonds, are well known in the art and described, for example, in The Peptides: Analysis, Synthesis, Biology, Vol. I., eds. Gross, et al., Academic Press, 1979, which is incorporated herein by reference in its entirety. An example F2 fragment is provided in Formula (E) (Pg is an amino protecting group, Z is defined herein). Additionally, protection of the amino group of amino acids using Boc or other amino protecting groups is well known in the art.
Figure US08283367-20121009-C00047
Compounds of Formula (E) that are amino acids or amino acid derivatives are available commercially or prepared by routine methods. For example, aza-serines can be prepared generally by the Hoffman Rearrangement (Hoffman's Reaction) using, for example, asparagine where the amide of the asparagine side chain is converted to an amine (which can be subsequently protected). Methods for carrying out Hoffman Rearrangements, such as for amino acids, are known in the art and also provided in the Examples below. Additionally aza-serines can be prepared as disclosed in Zhang, et al. J. Org. Chem., 1997, 62, 6918-6920, which is incorporated herein by reference in its entirety. F2 fragments can be obtained from commercial sources or made by methods known to one skilled in the art.
F3 Fragments
A further fragment (F3) can be coupled to the F2 fragment of the F2-F1 intermediate by any of various means such as by nucleophilic substitution or addition reactions where, for example, F2 contains a nucleophile (e.g., amine) and F3 contains an electrophile (e.g., CO) and optionally a leaving group (e.g., halo, hydroxy, alkoxy, alkylsulfonyl, arylsulfonyl, and the like). Example F3 fragments can have the formula HyCOOH. Coupling of HyCOOH to the F2-F1 intermediate can be carried out according to standard procedures for peptide bond formation to prepare compounds having the formula F3-F2-F1 where the F3and F2 fragments are coupled via an amide bond. Other coupling means are known in the art and are also suitable. F3 fragments can be obtained from commercial sources or made by methods known in the art.
F3-F2-F1 Product
The F3-F2-F1 product includes compounds of the invention and can also be derivatized to prepare additional compounds of the invention by routine methods in the art. For example, compounds of the invention where Z is —CH2NH2 can be prepared by removal of an amino protecting group such as benzyloxycarbonyl group (—C(═O)OCH2(C6H5)) which is attached to one of the nitrogens of the azaserine group (e.g., compounds of Formula (I) where Z is —CH2NHR1 and R1 is —C(═O)OCH2(C6H5)). Removal of the benzyloxycarbonyl group can be carried out by treatment with a reducing agent, such as a hydrogenation reagent. In some embodiments, the hydrogenation reagent contains H2 which is optionally used in the presence of a metal catalyst (e.g., Pd/C 10%). Hydrogenation can be further carried out in the presence of a protic acid such as HCl and in a suitable hydrogenation solvent containing, for example, an alcohol (e.g., methanol) and/or an ether solvent (e.g., 1,4-dioxane).
Certain compounds of the invention wherein Z is —CH2NHR1 can be prepared by removal of an R1 amino protecting group to form the corresponding deprotected amine (such as described above) followed by reaction with a reagent having the formula R1XL, (with the exception that R1 is not H; and XL is a leaving group such as halo or a sulfonic acid derivative; or wherein R1 and XL taken together represent, for example, a reactive alkyl, carbocyclyl or heterocarbocyclyl isocyanate, or an alkyl, carbocyclyl, heterocarbocyclyl sulphonylisocyanate).
Boronic Ester/Boronic Acid Conversion
Compounds of the invention containing boronic esters, such as pinanediol esters, can be hydrolyzed by any suitable means to prepare corresponding boronic acid (—B(OH)2) derivatives. Hydrolysis conditions can include contacting a boronic ester with excess acid, such as a protic acid like HCl.
Conversely, boronic acids can be esterified by contacting the acid compound (—B(OH)2) with an alcohol such as a diol for sufficient time to produce the corresponding ester. The esterification reaction can be acid or base catalyzed.
The invention will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of noncritical parameters which can be changed or modified to yield essentially the same results.
EXAMPLES Example A.1 Synthesis of intermediate (1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt Step 1: 2-(2-methylpropyl)-(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole
Figure US08283367-20121009-C00048
A mixture of (+)-pinanediol (23.9 g, 0.140 mol) and 2-methylpropylboronic acid (15 g, 0.147 mol) in diethyl ether (300 ml) was stirred at room temperature for 24 h. The mixture was dried over anhydrous sodium sulfate and purified by column chromatography (Silica gel 230-400 mesh), eluting with hexane:ethyl acetate 90:10 mixture. The product was obtained as a clear oil (32.6 g, 94% yield).
1H NMR (DMSO-d6): 4.28 (1H, dd, J=8.8 Hz, 2.0); 2.30 (1H, m); 2.18 (1H, m); 1.96 (1H, t, J=5.3); 1.86 (1H, m); 1.78 (1H, set, J=6.8); 1.68 (1H, m); 1.30 (3H, s); 1.25 (3H, s); 1.01 (1H, d); 0.9 (6H, d, J=6.6); 0.81 (3H, s); 0.69 (2H, m).
Step 2: 2-[(1S)-1-chloro-3-methylbutyl]-(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole
Figure US08283367-20121009-C00049
A solution of lithium diisopropylamide was prepared by addition of 10.0 M butyl lithium solution in hexane (25.4 ml, 0.254 mol) to a solution of diisopropylamine (35.7 ml, 0.254 mol) in dry tetrahydrofuran (60 ml), at −50° C., and allowing the temperature to rise to −30° C. This solution was transferred via canula into a solution of 2-(2-methylpropyl)-(3aS, 4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole of Step 1 (50 g, 0.212 mol) and CH2Cl2 (50 ml, 0.848 mol) in dry tetrahydrofuran (700 ml), while keeping the temperature below −70° C. A 1.0 M solution of dry zinc chloride in diethyl ether (339 ml, 0.339 mol) was then added over a 30 minutes period while keeping the internal temperature below −70° C. The reaction mixture was stirred at −78° C. for 3 hours, then allowed to warm to room temperature. After removal of the solvents by rotary evaporation the residue was partitioned between petroleum ether (1000 ml) and a 10% aqueous solution of ammonium chloride (800 ml). The aqueous layer was further extracted with petroleum ether (300 ml). The combined organic phases were dried over anhydrous sodium sulfate and concentrated. The product was obtained as a brown oil (59.0 g, 98% yield) containing about 9% mol/mol of starting material (1H-NMR), and was used in the subsequent step without further purification.
1H NMR (DMSO-d6): 4.43 (1H, dd, J=8.8, 1.8); 3.59 (1H, m); 2.33 (1H, m); 2.21 (1H, m); 2.01 (1H, m); 1.88 (1H, m); 1.84-1.55 (5H, m); 1.34 (3H, s); 1.26 (3H, s); 1.09 (1H, J=10.1); 0.9 (3H, d, J=6.8); 0.87 (3H, d, J=6.4); 0.82 (3H, s).
Step 3: N,N-Bis(trimethylsilyl)-(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine
Figure US08283367-20121009-C00050
A 1.0 M solution of lithium bis(trimethylsilyl)amide in tetrahydrofuran (189 ml, 0.189 mol) was added, over 30 minutes, to a solution of crude 2-[(1S)-1-chloro-3-methylbutyl]-(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole of Step 2 (59.0 g, 91% purity, 0.189 mol) in tetrahydrofuran (580 ml) while cooling at −78° C. The reaction mixture was allowed to slowly warm to room temperature overnight. The solvent was removed by rotary evaporation and the residue taken up with dry hexane (800 ml). The resulting suspension was stirred at room temperature for 2 hours, then the solid was removed by filtration on a celite cake, which was washed with dry hexane (3×100 ml). The filtrate was concentrated giving a satisfactorily pure product as a brown oil (79 g) in practically quantitative yield. The product was used for the subsequent step without further purification.
1H NMR (DMSO-d6): 4.33 (1H, dd, J=1.5 Hz, 8.6); 2.58 (1H, m); 2.29 (1H, m); 2.18 (1H, m); 1.95 (1H, t, J=5.9); 1.85 (1H, m); 1.9-1.55 (3H, m); 1.31 (3H, s); 1.24 (3H, s); 1.17 (1H, m); 1.01 (1H, d, J=10.6); 0.85 (3H, d, J=6.6), 0.83 (3H, d, J=6.6); 0.80 (3H, s); 0.08 (18H, s).
Step 4: (1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt
Figure US08283367-20121009-C00051
To a solution of crude N,N-bis(trimethylsilyl)-(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a, 5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine of Step 3 (79 g, 0.193 mol) in a mixture of dioxane (100 ml) and diethyl ether (200 ml), a 4 N solution of hydrogen chloride in dioxane (193 ml, 0.772 mol) was added, while cooling at 0° C. The mixture was then stirred at room temperature for 4 hours and concentrated. The residue was taken up with anhydrous hexane (500 ml) and a 2 M solution of hydrogen chloride in diethyl ether (48 ml, 0.096 mol) was added. The mixture was stirred at 0° C. for 1 hour, then concentrated. The residue was taken up with anhydrous hexane and the resulting suspension was stirred at room temperature overnight. The solid was collected by filtration and dried under vacuum affording 38.1 g of product (66% yield). A second crop (4.13 g, 7% yield) was obtained from the mother liquors.
1H NMR (DMSO-d6): 7.85 (3H, br); 4.45 (1H, dd, J=9.2 Hz); 2.78 (1H, m); 2.34 (1H, m); 2.21 (1H, m); 2.01 (1H, t, J=5.3); 1.89 (1H, m); 1.82-1.65 (2H, m); 1.49 (1H, m); 1.38 (3H, s); 1.27 (3H, s); 1.12 (1H, d, J=1.12); 0.87 (6H, d, J=6.6); 0.83 (3H, s).
Example A.2 Alternate Synthesis of Intermediate 2-(2-methylpropyl)-(3aS,4S,6S,7aR)-hexahydro-3a, 5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole Step 1: 2-(1-methylethoxy)-(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole
Figure US08283367-20121009-C00052
To a solution of (1S,2S,3R,5S)-(+)-pinanediol (50.0 g, 0.293 mol) in anhydrous tetrahydrofuran (350 ml) triisopropoxy borane was slowly added while stirring at 0° C. under nitrogen. After 2 h the solvent was removed by rotary evaporation. The oily residue was redissolved in hexane (150 ml) and the solution was filtered to remove a very small amount of a white solid. The filtrate was concentrated by rotary evaporation affording the product as a clear oil (62.6 g, 90% yield).
1H NMR (DMSO-d6): 4.31-4.20 (2H, m); 2.34-2.16 (2H, m); 1.96 (1H, t, J=5.5); 1.90-1.85 (1H, m); 1.74-1.67 (1H, m); 1.32 (3H, s); 1.31 (1H, d, J=7.6); 1.25 (3H, s); 1.14 (3H, d, J=6.1); 1.13 (3H, d, J=6.1); 0.81 (3H, s).
Step 2: 2-(2-methylpropyl)-(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole
Figure US08283367-20121009-C00053
A 2M solution of isobutyl magnesium bromide in diethyl ether (131.5 ml, 0.263 mol) was added dropwise, in 1 hour, to a solution of 2-(1-methylethoxy)-(3aS,4S,6S,7aR)-hexahydro-3a, 5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole obtained in Step 1 (62.6 g, 0.263 mol), in anhydrous tetrahydrofuran (330 ml) while stirring at −78° C., under nitrogen. The mixture was then allowed to warm to room temperature, then transferred in a mixture of 2N sulfuric acid (150 ml) and diisopropyl ether (250 ml). After stirring for 10 minutes, a saturated solution of NaCl was added (100 ml) and the layers were separated. The organic phase was washed with brine (100 ml), dried over sodium sulfate and concentrated. The residue was purified by column chromatography (silica gel) eluting with 5% diethyl ether in hexane. The product was obtained as a clear oil (38.45 g, 62% yield).
Example B.2 Preparation of Intermediate Carbamic acid 1,1-dimethylethyl ester, N-[(1S,2R)-1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2-hydroxypropyl]-
Figure US08283367-20121009-C00054
Boc-L-threonine (870 mg, 3.97 mmol, 1.2 eq.) was dissolved in DMF dry (30 ml) at r.t. To this solution, TBTU (N,N,N′,N′-tetramethyl-O-(benzotriazol-1-yl)uronium tetrafluoroborate; 1270 mg, 3.97 mmol, 1.2 eq.) was added and the mixture was cooled to 0°to 5° C. Then NMM (N-methylmorpholine, 0.9 ml, 8.27 mmol, 2.5 eq.) and (1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt of Example A.1 (1000 mg, 3.3 mmol, 1 eq.) were added. The mixture was stirred at r.t. for 16 h, then was extracted with ethyl acetate (100 ml) washed with the following solutions: citric acid 2% (50 ml), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated under reduced pressure to give 1290 mg of glassy solid. Yield 84.3%. M.p. 25°-30° C.
1H NMR (DMSO-d6): 8.88 (1H, br); 6.49 (1H, d, J=8.4 Hz); 4.88 (1H, d, J=5.8); 4.05 (1H, dd); 3.93 (1H, m); (1H, m); 2.51 (1H, m); 2.19 (1H, m); 2.01 (1H, m); 1.83 (1H, t, J=5.9), 1.78 (1H, m); 1.68 (1H, m); 1.62 (1H, m); 1.39 (9H, s); 1.34 (1H, d, J=10.0); 1.24 (3H, s); 1.22 (3H, s); 1.06 (3H, d, J=6.4); 0.85 (6H, d, J=6.4); 0.80 (3H, s)
Example B.3 Preparation of Intermediate Carbamic acid benzyl ester, N-[(1S,2R)-1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2-hydroxypropyl]
Figure US08283367-20121009-C00055
This intermediate was prepared using procedures analagous to those of Example B2 using appropriate starting materials.
M.p. 57-60° C. 1H NMR (DMSO-d6): 8.66 (1H, s); 7.40-7.29 (5H, m); 7.09 (1H, d, J=8.75); 5.06 (2H, s); 4.90 (1H, J=5.68); 4.11-3.99 (2H, m); 3.91-3.77 (1H, m); 2.58-2.53 (1H, m); 2.26-2.14 (1H, m); 2.07-1.97 (1H, s); 1.84 (1H, t, J=5.52); 1.81-1.75 (1H, m); 1.73-1.58 (2H, m); 1.33 (2H, d, J=10.1); 1.27-1.20 (7H, m); 1.06 (3H, t, J=6.27); 0.91-0.79 (9H, m).
Example B.4 Preparation of Intermediate (2S)-2-[(1,1-Dimethylethoxycarbonyl)amino]-3-[(4-methylbenzoyl)amino]propanamide, N-[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]-
Figure US08283367-20121009-C00056
(2S)-2-[(1,1-Dimethylethoxycarbonyl)amino]-3-[(4-methylbenzoyl)amino]-propanoic acid, (650 mg, 2 mmol, 1.2 eq.) of Example G.6, was dissolved in DMF dry (15 ml), under nitrogen, and TBTU (640 mg, 2 mmol, 1.2 eq.) was added at r.t. The mixture was cooled at 0°-5° C. with ice bath and NMM (0.55 ml, 5 mmol, 2.5 eq.) and (1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a, 5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt, (500 mg, 1.65 mmol, 1 eq.) of Example A.1, were added. The mixture was stirred overnight, poured in water (200 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (20 mL), sodium bicarbonate 2% (20 ml), NaCl 2% (20 ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated to give 740 mg of glassy solid (quantitative yield).
1H NMR (DMSO-d6) 8.76 (1H, br); 8.28 (1H, t, J=5.31 Hz); 7.71 (2H, d, J=7.9); 7.26 (2H, d, J=7.9); 6.97 (1H, d, J=8.0); 4.27 (1H, m); 4.07 (1H, dd, J=8.2, 1.5); 3.48 (2H, m), 2.58 (1H, m); 2.35 (3H, s); 2.19 (1H, m); 2.02 (1H, m); 1.83 (1H, t, J=4.9); 1.78 (1H, m); 1.62 (2H, m); 1.35 (12H, m); 1.24 (3H, s); 1.23 (3H, s); 0.82 (3H, d); 0.80 (3H, d); 0.78 (3H, s).
Example B.5 Preparation of Intermediate 2-S-[(1,1-Dimethylethoxycarbonyl)amino]-3-(hexanoylamino)-propionamide, N-[(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]
Figure US08283367-20121009-C00057
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid, (300 mg, 1 mmol, 1.2 eq.) of Example G.7 was dissolved in DMF dry (25 ml), under nitrogen, and TBTU (318 mg, 1 mmol, 1.2 eq.) was added at r.t. The mixture was cooled at 0°-5° C. with ice bath and NMM (0.27 ml, 2.47 mmol, 2.47 eq.) and (1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a, 5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt, (250 mg, 0.82 mmol, 1 eq.) of Example A.1, were added. The mixture was stirred 3 h, poured in water (150 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (50 mL), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated to give 450 mg of glassy solid. Yield quantitative.
Analytical data: 1H NMR (DMSO-d6). δH, 8.71 (1H, br d, J=2.6 Hz); 7.73 (1H, br t, J=5.9 Hz); 6.81 (1H, d, J=8.2); 4.10 (2H, m); 3.24 (2H, m); 2.56 (1H, m); 2.19 (1H, m); 2.03 (3H, m); 1.83 (1H, t, J=5.5); 1.78 (1H, m); 1.64 (2H, m); 1.47 (2H, m); 1.36 (9H, s); 1.4-1.15 (9H, m); 1.24 (3H, s); 1.21 (3H); 0.83 (9H, m); 0.79 (3H, s)
Example B.6 Preparation of Intermediate 2-S-[(1,1-Dimethylethoxycarbonyl)amino]-3-(4-fluorosulfonylamino)propionamide, N-[(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a, 5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-
Figure US08283367-20121009-C00058
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(4-fluorosulfonylamino)propionic acid, (1.39 g, 3.83 mmol, 1.2 eq.) of Example G.8, was dissolved in DMF dry (20 ml), under nitrogen, and TBTU (1.23 g, 3.83 mmol, 1.2 eq.) was added at r.t. The mixture was cooled at 0°-5° C. with ice bath and NMM (1 ml, 9.57 mmol, 3 eq.) and (1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a, 5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt, (0.96 g, 3.19 mmol, 1 eq.) of Example A.1, were added. The mixture was stirred 2 h, poured in water (200 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (50 mL), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated with diethyl ether to give 1.5 g of white solid. Yield 77%.
Analytical data:
1H NMR (DMSO-d6).
δH, 8.54 (1H, d, J=2.9 Hz); 7.91 (2H, m); 7.75 (1H, t, J=5.9); 7.50 (2H, t, J=8.8); 6.83 (1H, d, J=8.4); 4.19 (1H, br d, J=8.2); 4.14 (1H, m); 3.01 (2H, m); 2.69 (1H, m); 2.25 (1H, m); 2.09 (1H, m); 1.90 (1H, t, J=5.7); 1.85 (1H, m); 1.8-1.6 (2H, m); 1.5-1.2 (5H, m); 1.43 (9H, s); 1.29 (6H, s); 0.89 (6H, d, J=6.4); 0.86 (3H, s).
Example B.7 Preparation of Intermediate 2-S-[(1,1-Dimethylethoxycarbonyl)amino]-3-(3,4-dimethoxyphenylacetamido)propionamide, N-[(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a, 5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-
Figure US08283367-20121009-C00059
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(3,4-dimethoxyphenylacetamido)-propionic acid, (0.73 g, 1.90 mmol, 1.2 eq.) of Example G.9, was dissolved in DMF dry (20 ml), under nitrogen, and TBTU (0.61 g, 1.90 mmol, 1.2 eq.) was added at r.t. The mixture was cooled at 0°-5° C. with ice bath and NMM (0.52 ml, 4.7 mmol, 2.5 eq.) and (1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt, (0.47 g, 1.6 mmol, 1 eq.) of Example A.1, were added. The mixture was stirred 2 h, poured in water (200 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (50 mL), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated with diethyl ether to give 0.95 g of crude that was purified by silica gel chromatography (eluent ethyl acetate) to give 0.3 g of white foam. Yield 30%.
Analytical data: TLC silica gel (eluent ethyl acetate 100%, R.f.=0.50)
1H NMR (DMSO-d6).
δH, 8.69 (1H, d, J=2.6 Hz); 7.90 (1H, t, J=5.7); 6.85 (2H, m); 6.74 (1H, dd, J=1.5, 8.1); 6.85 (3H, m); 4.12 (2H, m); 3.73 (3H, s); 3.72 (3H, s); 3.34 (2H, s); 3.31 (2H, m); 2.58 (1H, m); 2.20 (1H, m); 2.03 (1H, m); 1.85 (1H, t, J=5.3); 1.79 (1H, m); 1.66 (2H, m); 1.38 (9H, s); 1.40-1.15 (3H, m); 1.25 (3H, s); 1.23 (3H, s); 0.83 (6H, d, J=6.6); 0.81 (3H, s).
Example B.8 Preparation of Intermediate 2-S-[(1,1-Dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionamide, N-[(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-
Figure US08283367-20121009-C00060
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid, (0.41 g, 1.26 mmol, 1.2 eq.) of Example G.10, was dissolved in DMF dry (20 ml), under nitrogen, and TBTU (0.40 g, 1.26 mmol, 1.2 eq.) was added at r.t. The mixture was cooled at 0°-5° C. with ice bath and NMM (0.346 ml, 3.15 mmol, 2.5 eq.) and (1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a, 5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt, (0.31 g, 1 mmol, 1 eq.) of Example A.1, were added. The mixture was stirred 2 h, poured in water (200 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (50 mL), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml). The organic solution was dried over sodium sulphate anhydrous, filtered and evaporated with diethyl ether (50 ml) to give 0.58 g of white solid.
Yield 96.6%.
Analytical data: TLC silica gel (eluent ethyl acetate 100%, R.f.=0.47), m.p. 128°-130° C.
1H NMR (DMSO-d6).
δH, 8.79 (1H, d, J=2.7 Hz); 8.69 (1H, s); 7.38 (2H, d, J=7.9); 7.22 (2H, t, J=8.1); 7.00 (1H, d, J=8.1); 6.90 (1H, t, J=7.3); 6.16 (1H, t, J=5.7); 4.12 (2H, m); 3.45 (1H, m); 3.17 (1H, m); 2.60 (1H, m); 2.21 (1H, m); 2.04 (1H, m); 1.85 (1H, t, J=5.3); 1.79 (1H, m); 1.66 (2H, m); 1.38 (9H, s); 1.40-1.15 (3H, m); 1.26 (3H, s); 1.23 (3H, s); 0.84 (6H, d, J=6.6); 0.81 (3H, s).
Example B.9 Synthesis of Further Intermediates
Following the procedures of Examples B.4-B.8, the following compounds can be prepared by reaction of (1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutylamine hydrochloride salt of Example A.1 and intermediates of Examples G.11, G.12 and G.13.
B.9.1 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3- (acetamido-)propionamide, N-[(1S)-1-[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl].
Figure US08283367-20121009-C00061
B.9.2 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(9- fluorenylmethyloxycarbamoyl)ethyl]-propionamide,N- [(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl].
Figure US08283367-20121009-C00062
B.9.3 2-S-[(1,1-dimethylethoxycarbonyl)amino]-2- [(pentylureido)ethyl]-N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-
Figure US08283367-20121009-C00063
B.9.4 2-S-[(1,1-dimethylethoxycarbonyl)amino]-2- (methanesolfonamido)ethyl]-N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-
Figure US08283367-20121009-C00064
B.9.5 2-S-[(1,1-dimethylethoxycarbonyl)amino]-2- [(ethoxycarbonylsuccinyl]-amide)ethyl]-N-[(1S)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-
Figure US08283367-20121009-C00065
B.9.6 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3- (benzyloxycarbamoyl)ethyl]-propionamide,N-[(1S)-1- [[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl].
Figure US08283367-20121009-C00066
B.9.7 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-[2-(1H- pyrazol)ethyl]-N-[(1S)-1-[[[(1R)-1-[(3aS,aS,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2- benzodioxaborol-2-yl-]- 3-methylbutyl]amino]carbonyl]
Figure US08283367-20121009-C00067
Example C.3 Preparation of Intermediate (2S,3R)-2-Amino-3-hydroxybutanamide, N-[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]-, hydrochloride salt
Figure US08283367-20121009-C00068
A 4 N solution of hydrogen chloride in dioxane was added to a solution of carbamic acid 1,1-dimethylethyl ester, N-[(1S,2R)-1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2-hydroxypropyl]- of Example B.2, in a mixture of dioxane and diethyl ether, while cooling to 0° C. The reaction mixture was allowed to warm to room temperature and stirred for several additional hours. The solvent was removed by rotary evaporation, the residue was treated with diethyl ether and the mixture was stirred at r.t. for several days. The resulting solid was collected by filtration affording pure product in good yield.
1H NMR (DMSO-d6) δH, 8.62 (1H, d, J=5.0 Hz); 8.17 (3H, d, J=3.5); 4.28 (1H, dd, J=8.8, 1.8); 3.78 (1H, m); 3.52 (1H, m); 3.00 (1H, m); 2.28 (1H, m); 2.10 (1H, m); 1.92 (1H, t, J=5.7); 1.84 (1H, m); 1.75-1.62 (2H, m); 1.43 (1H, m); 1.31 (3H, s); 1.25 (3H, s); 1.22 (1H, d, J=10.6); 1.14 (3H, d, J=6.2); 0.88 (3H, d, J=6.4); 0.86 (3H, d, J=6.4); 0.81 (3H, s)
Example C.4 Preparation of Intermediate (2S)-2-Amino-3-[(4-methylbenzoyl)amino]propanamide, N-[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]-, hydrochloride salt
Figure US08283367-20121009-C00069
(2S)-2-[(1,1-Dimethylethoxycarbonyl)amino]-3-[(4-methylbenzoyl)-amino]-propanamide, N-[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]-, Example B.4, (740 mg, 1.65 mmol, 1 eq.), was dissolved in 1,4-dioxane (20 ml). To this solution, HCl 4N in 1,4-dioxane (5 ml, 19.8 mmol, 12 eq.) was added and the solution stirred overnight at r.t. The solvent was removed under reduced pressure to give 800 mg of a glassy solid (quantitative yield).
1H NMR (DMSO-d6) 8.63 (1H, d, J=5.5 Hz); 8.38 (1H, t, J=8.4 Hz); 8.34 (3H, br); 7.80 (2H, t, J=8.2); 7.28 (2H, d, J=8.2 Hz); 4.15 (1H, dd, J=8.8, 1.8); 4.02 (1H, br); 3.66 (1H, m); 3.55 (1H, m); 2.99 (1H, m); 2.35 (3H, s); 2.19 (1H, m); 2.06 (1H, m); 1.86 (1H, t, J=5.7); 1.80 (1H, m); 1.64 (2H, m); 1.41 (1H, m); 1.33-1.19 (2H, m); 1.27 (3H, s), 1.21 (3H, s); 1.16 (1H, d, J=10.6); 0.82 (3H, d); 0.80 (3H, d); 0.78 (3H, s).
Example C.5 2-S-amino-3-(hexanoylamino)-propionamide, N-[(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a, 5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl], hydrochloride salt
Figure US08283367-20121009-C00070
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionamide, N-[(1S) -1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl], of Example B.5, (450 mg, 0.8 mmol, 1 eq.), was dissolved in 1,4-dioxane (15 ml). To this solution, HCl 4N in 1,4-dioxane (2.45 ml, 0.98 mmol, 12 eq.) was added and the solution stirred overnight at r.t. The solvent was removed under reduced pressure to give 400 mg of a glassy solid. Yield quantitative.
Analytical data: 1H NMR (DMSO-d6).
δH, 8.54 (1H, d, J=5.3 Hz); 8.18 (3H, br); 7.74 (1H, t, J=5.7); 4.29 (1H, dd, J=1.8, 8.8); 3.83 (1H, m); 3.40 (2H, m); 3.00 (1H, m); 2.29 (1H, m); 2.11 (1H, m); 2.08 (2H, t, J=7.5); 1.93 (1H, t, J=5.5); 1.84 (1H, m); 1.75-1.15 (11H, m); 1.32 (3H, s); 1.24 (3H, s); 0.86 (3H, d, J=6.6); 0.84 (3H, d, J=6.6); 0.81 (3H, s).
Example C.6 Preparation of Intermediate 2-S-amino-3-(4-fluorosulfonylamino)propionamide, N-[(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl], hydrochloride salt
Figure US08283367-20121009-C00071
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(4-fluorosulfonylamino)propionamide, N-[(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl], of Example B.6, (0.7 g, 1.14 mmol, 1 eq.), was dissolved in 1,4-dioxane (20 ml). To this solution, HCl 4N in 1,4-dioxane (3.4 ml, 13.68 mmol, 12 eq.) was added and the solution stirred overnight at r.t. The solvent was removed under reduced pressure to give 440 mg of a white solid. Yield 71%. Analytical data:
1H NMR (DMSO-d6).
δH, 8.54 (1H, d, J=5.5 Hz); 8.26 (3H, br); 7.89 (3H, m); 7.48 (3H, t, J=8.8); 4.26 (1H, dd, J=1.3, 8.6); 3.84 (1H, m); 3.06 (2H, m); 2.97 (1H, m); 2.25 (1H, m); 2.03 (1H, m); 1.83 (2H, m); 1.64 (2H, m); 1.42 (1H, m); 1.35-1.15 (3H, m); 1.28 (3H, s); 1.22 (3H, s); 1.11 (1H, d, J=10.8); 0.85 (6H, m); 0.80 (3H, s).
Example C.7 2-S-amino-3-(3,4-dimethoxyphenylacetamido)propionamide, N-[(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl], hydrochloride salt
Figure US08283367-20121009-C00072
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(3,4-dimethoxyphenylacetamido)-propionamide, N-[(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl], of Example B.7, (0.3 g, 0.47 mmol, 1 eq.), was dissolved in 1,4-dioxane (20 ml). To this solution, HCl 4N in 1,4-dioxane (1.43 ml, 5.71 mmol, 12 eq.) was added and the solution stirred overnight at r.t. The solvent was removed under reduced pressure, diethyl ether was added and evaporated to give 230 mg of a white solid. Yield 85%.
Analytical data:
1H NMR (DMSO-d6).
δH, 8.57 (1H, br); 8.12 (3H, br); 7.91 (1H, t, J=5.7 Hz); 6.86 (2H, m); 6.76 (1H, dd, J=1.8, 8.2); 4.26 (1H, br d, J=7.3); 3.82 (1H, m); 3.72 (3H, s); 3.71 (3H, s); 3.36 (2H, s); 3.34 (2H, m); 2.99 (1H, m); 2.26 (1H, m); 2.10 (1H, m); 1.92 (1H, t, J=5.3); 1.83 (1H, m); 1.67 (2H, m); 1.45-1.15 (3H, m); 1.31 (3H, s); 1.23 (3H, s); 0.86 (3H, d, J=6.6); 0.84 (3H, d, J=6.6); 0.80 (3H, s).
Example C.8 Preparation of Intermediate 2-S-amino-3-(3-phenyl-ureido)-propionamide, N-[(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl], hydrochloride salt
Figure US08283367-20121009-C00073
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionamide, N-[(1S) -1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl], of Example B.8, (0.58 g, 0.1 mmol, 1 eq.), was dissolved in 1,4-dioxane (25 ml). To this solution, HCl 4N in 1,4-dioxane (3 ml, 12.1 mmol, 12 eq.) was added and the solution stirred overnight at r.t. The solvent was removed under reduced pressure, diethyl ether was added and evaporated to give 0.52 g of desired product. Yield 100%.
Analytical data:
1H NMR (DMSO-d6).
δH, 8.82 (1H, s); 8.59 (1H, d, J=5.7 Hz); 8.18 (3H, br); 7.40 (2H, d, J=7.9); 7.22 (2H, t, J=8.1); 6.90 (1H, t, J=7.3); 6.31 (1H, t, J=5.7); 4.26 (1H, dd, J=1.5, 8.6); 3.89 (1H, m); 3.48 (1H, m); 3.36 (1H, m); 3.01 (1H, m); 2.24 (1H, m); 2.10 (1H, m); 1.92 (1H, t, J=5.3); 1.82 (1H, m); 1.67 (2H, m); 1.50-1.15 (3H, m); 1.31 (3H, s); 1.21 (3H, s); 0.85 (3H, d, J=6.6); 0.84 (3H, d, J=6.6); 0.79 (3H, s).
Example C.9 Synthesis of Further Intermediates
Following the procedures of Examples C.4-C.8, the following compounds can be prepared starting from intermediates of Example B.9.
C.9.1 2-S-amino-3-(acetamido)-propionamide,N-[(1S)-1-[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl], HCl salt.
Figure US08283367-20121009-C00074
C.9.2 2-S-amino-3-(9-fluorenylmethyloxycarbamoyl)- propionamide,N-[(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl], HCl salt.
Figure US08283367-20121009-C00075
C.9.3 2-S-amino-3-(pentylureido)-propionamide, N-[(1S)-1- [[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl], HCl salt.
Figure US08283367-20121009-C00076
C.9.4 2-S-amino-3-(methanesolfonamido)-propionamide,N- [(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl], HCl salt.
Figure US08283367-20121009-C00077
C.9.5 2-S-amino-3-[(ethoxycarbonylsuccinyl]-amide)ethyl]-)- propionamide, N-[(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)- hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2- benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl], HCl salt.
Figure US08283367-20121009-C00078
C.9.6 2-S-amino-3-(benzyloxycarbamoyl)-propionamide,N- [(1S)-1-[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl], HCl salt.
Figure US08283367-20121009-C00079
C.9.7 3-[2-(1H-pyrazol)ethyl]-N-[(1S)-1-[[[(1R)-1- [(3aS,aS,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl-]- 3-methylbutyl]amino]carbonyl] HCl salt.
Figure US08283367-20121009-C00080
Example G.5 Preparation of Intermediate 3-Amino-2-S-[(1,1-dimethylethoxycarbonyl)amino]-propionic acid, benzyl ester
Figure US08283367-20121009-C00081
Step 1: N-tert-butoxycarbonyl-L-asparagine [Commercially available]
Figure US08283367-20121009-C00082
L-asparagine (15 g, 0.113 mol, 1 eq.) and sodium carbonate (12 g, 0.113 mol) were dissolved in water (225 ml) and 1,4-dioxane (225 ml) at r.t. To this solution, di-tert-butyl-dicarbonate (30 g, 0.137 mol, 1.2 eq.) was added and the mixture was stirred overnight. The solvent was evaporated under reduced pressure until 1,4-dioxane was distilled and the pH adjusted to 2 with HCl 37% to give a white solid that was filtered, washed with water and dried. Yield 91%. 24 g.
Analytical data: m.p. 175° C.-180° C. (lit. 175° C.).
1H NMR (DMSO-d6) 12.5 (1H, br); 7.31 (1H, br); 6.91 (1H, br); 6.87 (1H, d, J=8.4 Hz); 4.23 (1H, q, J=7.7 Hz); 2.56-2.36 (2H, m); 1.38 (9H, s).
Step 2: N-[(1,1-dimethylethoxycarbonyl)amino]-L-asparagine, benzyl ester
Figure US08283367-20121009-C00083
The compound was prepared according to Bioorg. Med. Chem., 6 (1998)1185-1208. N-[(1,1-dimethylethoxycarbonyl)amino]-L-asparagine (20.7 g, 89.1 mmol, 1 eq.), of Step 1, was dissolved in methanol (500 ml) and cesium carbonate (15.97 g, 49 mmol, 0.55 eq.) was added. The solvent was evaporated to give a white solid that was dissolved in N,N-dimethylformamide (200 ml). To the suspension, benzyl bromide (11.6 ml, 98 mmol, 1.1 eq.) was added dropwise and the mixture was stirred overnight. The solvent was reduced under reduced pressure, water (300 ml) was added and the mixture extracted with ethyl acetate (200 ml), washed with brine (50 ml) and the solvent removed under reduced pressure to give a crude that was suspended in n-hexane (160 ml), filtered and dried under vacuum to give 14.68 g of white solid. Yield 51%.
Analytical data: m.p. 113°-115° C.
1H NMR (DMSO-d6) 7.35 (6H, m); 7.13 (1H, d, J=7.9 Hz); 6.94 (1H, br s); 5.10 (2H, s); 4.39 (1H, q, J=7.4 Hz); 2.6-2.4 (2H, m); 2.03 (2H, t, J=7.3); 1.37 (9H, s).
Step 3: 3-Amino-2-S-[(1,1-dimethylethoxycarbonyl)amino]-propionic acid, benzyl ester
Figure US08283367-20121009-C00084
N-[(1,1-dimethylethoxycarbonyl)amino]-L-asparagine, benzyl ester, (2 g, 6.3 mmol, 1 eq.), of Step 2, was dissolved in acetonitrile (80 ml) and water (80 ml). The solution was cooled to 0°-5° C. and iodobenzene diacetate (3 g, 9.3 mmol, 1.5 eq.) was added portionwise. The mixture was stirred at 0° C. for 30′, then at r.t. for 4 h. The organic solvent was removed under vacuum, diethyl ether and HCl 1N were added. The acqueous layer was separated and extracted with dichloromethane (100 ml) and sodium bicarbonate (3.5 g). The organic solvent was dried over sodium sulphate anhydrous, evaporated under reduced pressure to give 0.65 g of colourless oil. Yield 36%
Analytical data:
1H NMR (DMSO-d6) 7.45-7.20 (7H, m); 7.20 (1H, d, J=7.7 Hz); 5.13 (2H, AB q, J=12.8); 4.01 (1H, m); 2.80 (2H, m); 1.38 (9H, s).
Example G.6 Preparation of Intermediate (2S)-2-[(1,1-dimethylethoxycarbonyl)amino]-3-[(4-methylbenzoyl)amino]propanoic acid
Figure US08283367-20121009-C00085
Step 1: 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(4-methylbenzoylamino)propionic acid, benzyl ester
Figure US08283367-20121009-C00086
3-Amino-2-S-[(1,1-dimethylethoxycarbonyl)amino]-propionic acid, benzyl ester, (690 mg, 2.34 mmol, 1 eq.), of Example G.5, was dissolved in DMF dry (20 ml) and TBTU (900 mg, 2.98 mmol, 1.2 eq.) was added. The mixture was stirred at r.t. for 10′, cooled to 0°-5° C. with ice bath and NMM (0.51 ml, 4.68 mmol, 2 eq.) and 4-methyl benzoic acid (380 mg, 2.81 mmol, 1.2 eq.) were added. The mixture was stirred at r.t. for 3 h, poured in water (100 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with a solution of citric acid 2% (50 ml), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml), dried over sodium sulphate anhydrous and evaporated at reduced pressure to give 1 g of oil. Yield quantitative.
Analytical data:
1H NMR (DMSO-d6) 8.46 (1H, br t, J=5.7 Hz); 7.70 (2H, d, J=8.0); 7.35-7.2 (8H, m); 5.07 (2H, s); 4.29 (1H, m); 3.67 (1H, m); 3.58 (1H, m); 2.36 (3H, s); 1.37 (9H, s).
Step 2: (2S)-2-[(1,1-dimethylethoxycarbonyl)amino]-3-(4-methylbenzoylamino)propionic acid
Figure US08283367-20121009-C00087
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(4-methylbenzoylamino)-propionic acid, benzyl ester, (930 mg, 2.25 mmol), of Step 1, was dissolved in methanol (25 ml) and Pd/C 10% (90 mg) was added. The mixture was hydrogenated at atmospheric pressure for 1 h. Pd/C was filtered and the solution was evaporated under reduced pressure to give 650 mg of white foam. Yield 86%. Analytical data:
1H NMR (DMSO-d6): 12.5 (1H, br); 8.40 (1H, t, J=5.7 Hz); 7.71 (2H, d, J=8.05 Hz), 7.27 (2H, d, J=8.05 Hz); 7.09 (1H, d, J=7.9), 4.17 (1H, m); 3.57 (2H, m); 2.35 (3H, s); 1.37 (9H, m).
Example G.7 Preparation of Intermediate 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid
Figure US08283367-20121009-C00088
Step 1: 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid, benzyl ester
Figure US08283367-20121009-C00089
Hexanoic acid (450 mg, 3.87 mmol, 1.2 eq.) was dissolved in DMF dry (15 ml) and TBTU (1.24 g, 3.87 mmol, 1.2 eq.) was added, the mixture was stirred at r.t. for 20′, then was cooled to 0°-5° C. with ice bath. 3-amino-2-S-[(1,1-dimethylethoxycarbonyl)amino]propionic acid, benzyl ester, (950 mg, 3.22 mmol, 1 eq.), of Example G.5, and NMM (1.06 ml, 9.61 mmol, 2.5 eq.) were added. The mixture was stirred at r.t. overnight, poured in water (150 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with a solution of citric acid 2% (50 ml), sodium bicarbonate 2% (50 ml), NaCl 2% (50 ml), dried over sodium sulphate anhydrous and evaporated at reduced pressure to give a crude that was purified by silica gel column chromatography (eluent: n-hexane/ethyl acetate 2/1, R.f=0.52) 0.5 g of colourless oil. Yield 40%.
Analytical data:
1H NMR (DMSO-d6).
δH, 7.87 (1H, br t, J=6.2 Hz); 7.35 (5H, m); 7.14 (1H, d, J=8.2); 5.07 (2H, s); 4.14 (1H, m); 3.37 (2H, m); 2.00 (2H, t, J=7.1); 1.43 (2H, m); 1.36 (9H, s); 1.3-1.1 (4H, m); 0.83 (3H, t, J=7.1 Hz)
Step 2: 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid
Figure US08283367-20121009-C00090
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(hexanoylamino)propionic acid, benzyl ester (500 mg, 1.27 mmol), of Step 1, was dissolved in methanol (15 ml) and Pd/C 10% (50 mg) was added. The mixture was hydrogenated at atmospheric pressure for 1 h. Pd/C was filtered and the solution was evaporated under reduced pressure to give 300 mg of white solid. Yield 78%.
Analytical data: m.p. 123°-125° C.
1H NMR (DMSO-d6).
δH, 12.6 (1H, br); 7.84 (1H, br t); 6.87 (1H, d, J=7.5 Hz); 4.00 (1H, m); 3.32 (2H, m); 2.04 (2H, t, J=7.5); 1.47 (2H, m); 1.38 (9H, s); 1.3-1.1 (4H, m); 0.85 (3H, t, J=7.1 Hz)
Example G.8 Preparation of Intermediate 2-S-tert-butoxycarbonylamino-3-(4-fluorosulfonylamino)propionic acid
Figure US08283367-20121009-C00091
Step 1: 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(4-fluorosulfonylamino)propionic acid, benzyl ester
Figure US08283367-20121009-C00092
3-Amino-2-S-[(1,1-dimethylethoxycarbonyl)amino]propionic acid, benzyl ester (1.25 g, 4.24 mmol, 1 eq.), of Example G.5, was dissolved in dichloromethane dry (20 ml) and the solution was cooled to 0°-5° C., under nitrogen. TEA (0.65 ml, 4.67 mmol., 1.1 eq.) and 4-fluoro-sulfonylchloride (0.9 g, 4.67 mmol., 1.1 eq.) in dichloromethane dry (10 ml) were added. The mixture was stirred at r.t. for 1 h, evaporated under reduced pressure and diethyl ether (25 ml) was added and a white solid was obtained that was filtered and dried under vacuum to give 1.89 g of product. Yield 99%.
Analytical data: m.p. 105°-107° C. TLC silica gel (eluent: n-hexane/ethyl acetate 1/1, Rf=0.55).
1H NMR (DMSO-d6).
δH, 7.91 (1H, t, J=6.2 Hz); 7.85 (2H, dd, J=5.3, 8.8); 7.43 (2H, t, J=8.8); 7.35 (5H, m); 7.15 (1H, d, J=8.2); 5.09 (2H, s); 4.14 (1H, m); 3.10 (2H, m); 1.36 (9H, s).
Step 2: 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(4-fluorosulfonylamino)propionic acid
Figure US08283367-20121009-C00093
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(4-fluorosulfonylamino)propionic acid, benzyl ester (1.8 g, 3.98 mmol.), of Step 1, was dissolved in methanol (30 ml) and Pd/C 10% (180 mg) was added. The mixture was hydrogenated at atmospheric pressure for 1 h. Pd/C was filtered and the solution was evaporated under reduced pressure to give 1.39 g of colourless oil. Yield 97%.
Analytical data:
1H NMR (DMSO-d6).
δH, 12.7 (1H, br); 7.83 (2H, dd, J=5.3, 8.8); 7.78 (1H, br t, J=5.5); 7.42 (2H, t, J=8.8); 6.87 (1H, d, J=8.6); 3.99 (1H, m); 3.03 (2H, m); 1.36 (9H, s).
Example G.9 Preparation of Intermediate 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(3,4-dimethoxyphenylacetamido)-propionic acid
Figure US08283367-20121009-C00094
Step 1: 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(3,4-dimethoxyphenylacetamido)-propionic acid, benzyl ester
Figure US08283367-20121009-C00095
3,4-Dimethoxy-phenylacetic acid (720 mg, 3.66 mmol, 1.2 eq.) was dissolved in DMF dry (20 ml) and TBTU (1.17 g, 3.66 mmol, 1.2 eq.) was added, the mixture was stirred at r.t. for 20′, then was cooled to 0°-5° C. with ice bath. 3-amino-2-S-tert-butoxycarbonylamino-propionic acid, benzyl ester (0.9 g, 3.05 mmol, 1 eq.), of Example G.5, and NMM (1.0 ml, 9.15 mmol, 2.5 eq.) were added. The mixture was stirred at 0° C. for 2 h, then poured in water (200 ml) and extracted with ethyl acetate (100 ml). The organic layer was washed with the following solutions: citric acid 2% (20 ml), sodium bicarbonate 2% (20 ml), NaCl 2% (20 ml), dried over sodium sulphate anhydrous and evaporated at reduced pressure to give a crude that was purified by silica gel chromatography (eluent: n-hexane/ethyl acetate 1/1, R.f=0.57) to give 1 g of colourless oil. Yield 69%.
Analytical data: 1H NMR (DMSO-d6). δH, 8.02 (1H, t, J=5.7 Hz); 7.34 (5H, m); 7.17 (1H, d, J=7.7); 6.82 (2H, m); 6.71 (1H, dd, J=1.5, 8.2); 5.03 (2H, s); 4.14 (1H, m); 3.71 (3H, s); 3.69 (3H, s); 3.39 (2H, m); 1.36 (9H, s).
Step 2: 2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(3,4-dimethoxyphenylacetamido)-propionic acid
Figure US08283367-20121009-C00096
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(3,4-dimethoxyphenylacetamido)-propionic acid, benzyl ester (1 g, 2.1 mmol.), of Step 1, was dissolved in methanol (30 ml) and Pd/C 10% (10 mg) was added. The mixture was hydrogenated at atmospheric pressure for 1 h. Pd/C was filtered and the solution was evaporated under reduced pressure to give 0.73 g of white foam. Yield 91%.
Analytical data: 1H NMR (DMSO-d6). δH, 12.7 (1H, br); 8.06 (1H, t, J=5.9 Hz); 7.00 (1H, d, J=8.05); 6.91 (2H, m); 6.80 (1H, dd, J=1.5, 8.4); 4.08 (1H, m); 3.80 (3H, s); 3.78 (3H, s); 3.5-3.3 (2H, m); 1.36 (9H, s).
Example G.10 Preparation of Intermediate 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid
Figure US08283367-20121009-C00097
Step 1: 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid, benzyl ester
Figure US08283367-20121009-C00098
3-Amino-2-S-[(1,1-dimethylethoxycarbonyl)amino]propionic acid, benzyl ester (1.14 g, 3.87 mmol, 1 eq.), of Example G.5, was dissolved in dichloromethane (20 ml) at r.t. The solution was cooled to 0°-5° C. and phenyl isocyanate (0.42 ml, 3.87 mmol, 1 eq.) in dichlorometane (5 ml) was added dropwise. The solution was stirred at r.t. for 1 h, evaporated under reduced pressure and purified by silica gel chromatography (eluent n-hexane/ethyl acetate 1/1) to give 0.71 g of glassy solid that was suspended in diethyl ether to give a white solid. Yield 44%. Analytical data: TLC silica gel (eluent n-hexane/ethyl acetate 1/1 R.f.=0.44), m.p. 48°-50° C.
1H NMR (DMSO-d6). δH, 8.68 (1H, s); 7.4-7.27 (8H, m); 7.22 (2H, t, J=8.2 Hz); 6.90 (1H, t, J=7.3); 6.26 (1H, t, J=5.7); 5.11 (2H, s); 4.12 (1H, m); 3.58 (1H, m); 3.28 (1H, m); 1.38 (9H, s).
Step 2: 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid
Figure US08283367-20121009-C00099
2-S-[(1,1-dimethylethoxycarbonyl)amino]-3-(3-phenylureido)propionic acid, benzyl ester (0.7 g, 1.7 mmol.), of Step 1, was dissolved in methanol (25 ml) and Pd/C 10% (70 mg) was added. The mixture was hydrogenated at atmospheric pressure for 1 h. Pd/C was filtered and the solution was evaporated under reduced pressure to give 0.47 g of desired product.
Yield 87%.
Analytical data: 1H NMR (DMSO-d6). δH, 12.6 (1H, br); 8.66 (1H, s); 7.37 (2H, d, J=8.1 Hz); 7.21 (2H, t, J=7.50); 7.08 (1H, d, J=7.9); 6.89 (1H, t, J=7.3); 6.21 (1H, t, J=5.9); 3.98 (1H, m); 3.54 (1H, m); 3.22 (1H, m); 1.38 (9H, s).
Example G.11 Synthesis of Further Intermediates
The following compounds can be prepared starting from 3-amino-2-S-[(1,1-dimethylethoxycarbonyl)amino]propionic acid, benzyl ester of Example G.5, with the methods described in Step 1 and Step 2 of Examples G.6-G.10.
G.11.1 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(acetamido)propionic acid.
Figure US08283367-20121009-C00100
G.11.2 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(9- fluorenylmethyloxycarbamoyl))propionic acid.
Figure US08283367-20121009-C00101
G.11.3 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(3- pentylureido)propionic acid.
Figure US08283367-20121009-C00102
G.11.4 2-[(1,1-dimethylethoxycarbonyl)amino]-3- (methanesolfonamido)propionic acid.
Figure US08283367-20121009-C00103
G.11.5 2-[(1,1-dimethylethoxycarbonyl)amino]-3- [(ethoxycarbonylsuccinyl]-amide)ethyl]-propionic acid.
Figure US08283367-20121009-C00104
Example G.12 Preparation of Intermediate 2-[(1,1-Dimethylethoxycarbonyl)amino]-3-(3-benzyloxycarbonylamino)propionic acid
Figure US08283367-20121009-C00105
Step 1: N2-(tert-butoxycarbonyl)-L-2,3-diaminopropionic acid
Figure US08283367-20121009-C00106
N-tert-butoxycarbonyl-L-asparagine, from step 1 of Example G.5 or commercially available, (8 g, 0.034 mol, 1 eq.) was suspended in ethyl acetate (72 ml), acetonitrile (72 ml) and water (36 ml), and iodobenzenediacetate (13.3 g, 0.041 mol, 1.2 eq.) was added at 5° C. The mixture was stirred at 10°-25° C. for 3-4 h, then a white solid came off. The solid was filtered, washed with diethyl ether and dried under vacuum to give a white powder. Yield 57%. 4 g.
Analytical data: m.p. 210° C.-211° C. Silica gel (dichloromethane/methanol/acetic acid 5/3/1) Rf 0.5. 1H NMR (DMSO-d6) 4.15 (1H, t); 3.15 (2H, m); 1.45 (9H, s);
Step 2: 2-[(1,1-dimethylethoxycarbonyl)amino]-3-(3-benzyloxycarbonylamino)propionic acid
Figure US08283367-20121009-C00107
N2-(tert-Butoxycarbonyl)-L-2,3-diaminopropionic acid, from step 1, (3.8 g, 0.018 mol, 1 eq.) was dissolved in aqueous sodium carbonate 10% (2.2 eq.) at 25° C. and 1,4-dioxane (38 ml). To this solution, benzylchloroformate (3 ml, 0.020 mol, 1.1 eq.) was added dropwise and the solution was stirred at 25° C. for 3 h. At the end of the reaction, the mixture was poured in water (100 ml) and washed with diethyl ether (100 ml). To the aqueous solution, HCl 37% (6 ml) was added till pH 2 and the obtained mixture was extracted with Ethyl Acetate (100 ml). The organic layer was separated, washed with brine and dried over sodium sulfate anhydrous. The solvent was removed under reduced pressure to give a colourless oil that under vacuum gave a white foam. Yield 93%, 5.9 g.
Analytical data: silica gel (dichloromethane/methanol/acetic acid 5/3/1) Rf 1.
1H NMR (DMSO-d6) 12.6 (1H br s); 7.35 (5H m); 6.94 (1H, d); 5 (2H, s); 4.1 (2H, m); 1.4 (9H, s).
Example G.13 Preparation of Intermediate 2-(tert-butoxycarbonilamino)-3-pyrazol-1-yl-propionic acid
Figure US08283367-20121009-C00108
The intermediate was prepared according to the procedure described in Vederas, J. Am. Chem. Soc., 1985, 107, 7105-7109.
Example H.1 Preparation of 6-phenyl-pyrazine-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2-hydroxypropyl]
Figure US08283367-20121009-C00109
6-Phenyl-pyrazine-2-carboxylic acid (192 mg, 0.96 mmol) and TBTU (310 mg, 0.96 mmol) were dissolved in dry DMF (4 ml), under a nitrogen atmosphere. The reaction mixture was cooled to 0° C. and N-methyl-morpholine (2.29 ml, 2.61 mmol) was added. The mixture was stirred for 30 minutes, then (2S,3R)-2-amino-3-hydroxybutanamide, N-[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]-, hydrochloride salt of Example C.3 (350 mg, 0.87 mmol) was added and the mixture was allowed to warm to room temperature. After 3 h the reaction mixture was diluted with ethyl acetate (100 ml) then washed with water (50 ml), 2% citric acid (50 ml), 2% NaHCO3 (50 ml) and brine (50 ml). The organic phase was dried over anhydrous sodium sulfate, filtered and evaporated under reduced pressure. The residue was purified by elution on a SPE-SI normal phase cartridge (20 g SiO2) using first a 1:1 hexane:ethyl acetate mixture, then ethyl acetate. The title compound was obtained as a white foam (333 mg, 70 yield).
1H NMR (DMSO-d6): δ 9.39 (1H, s); 9.25 (1H, s); 8.96-8.92 (1H, bs); 8.52 (1H, d, J=8.5); 8.29-8.21 (2H, m); 7.64-7.55 (3H, m); 5.27 (1H, d, J=5.0); 5.54-5.49 (1H, m); 4.19-4.10 (2H, m); 2.66-2.59 (1H, m); 2.25-2.15 (1H, m); 2.07-2.00 (1H, m); 1.80-1.60 (3H, m); 1.35-1.26 (3H, m); 1.25 (3H, s); 1.22 (3H, s); 1.11 (3H, d, J=6.1); 0.87-0.79 (9H, m).
Example H.2 Preparation of Further Compounds of the Invention
Further compounds of the invention are listed below which can be made according to procedures analogous to those described for Example H.1 using the appropriate carboxylic acid such as described in Example M.1. Those compounds listed below which are characterized by NMR data were actually prepared.
Ex
# Structure Chemical Name and Analytical Data
H.2.1
Figure US08283367-20121009-C00110
Pyridine-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]
H.2.2
Figure US08283367-20121009-C00111
Pyridine-3-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]
H.2.3
Figure US08283367-20121009-C00112
Quinoline-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H NMR (DMSO-d6): 9.00-8.95 (1 H, bs); 8.76 (1 H, d, J = 8.4); 8.61 (1 H, d, J = 8.4); 8.22-8.10 (3 H, m); 7.90 (1 H, t, J = 7.6); 7.75 (1 H, t, J = 7.5); 5.28 (1 H, d, J = 4.8); 4.53-5.48 (1 H, m); 4.18-4.10 (2 H, m); 2.67- 2.60 (1 H, m); 2.26-2.18 (1 H, m); 2.08-2.00 (1 H, m); 1.86-1.78 (2 H, m); 1.74-1.60 (2 H, m); 1.36-1.26 (2 H, m); 1.24 (3 H, s); 1.21 (3 H, s); 1.14 (3 H, d, J = 5.9); 0.86-0.79 (9 H, m)
H.2.4
Figure US08283367-20121009-C00113
Quinoxaline-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H-NMR (DMSO-d6): 9.57 (1 H, s); 8.97 (1 H, br. s), 8.67 (1 H, d, J = 8.6); 8.35-8.26 (2 H, m); 8.11-8.05 (2 H, m); 5.34 (1 H, d, J = 5.0); 4.58 (1 H, dd, J = 8.4, 4.4); 4.23-4.16 (2 H, m); 2.75-2.68 (1 H, m); 2.31-2.22 (1 H, m); 2.12-2.04 (1 H, m); 1.90 (1 H, t, J = 5.5); 1.87-1.81 (1 H, m); 1.77-1.65 (2 H, m); 1.44-1.24 (3 H, m); 1.30 (3 H, s); 1.27 (3 H, s); 1.21 (3 H, d, J = 6.3); 0.89 (6 H, d, J = 6.5); 0.86 (3 H, s).
H.2.5
Figure US08283367-20121009-C00114
5-Phenyl-pyrazine-2-carboxamide, N-[(1S,2R)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H NMR (DMSO-d6): 9.52 (1 H, s); 9.18 (1 H, s); 8.91 (1 H, s); 8.66 (1 H, d, J = 8.5); 8.29-8.24 (2 H, m); 7.65- 7.57 (3 H, m); 5.27 (1 H, d, J = 4.7); 4.51-4.45 (1 H, m); 4.16-4.09 (2 H, m); 2.66-2.59 (1 H, m); 2.25-2.15 (1 H, m); 2.07-1.99 (1 H, m); 1.80-1.70 (2 H, m); 1.65-1.60 (2 H, m); 1.40-1.30 (3 H, m); 1.25 (3 H, s); 1.22 (3 H, s); 1.13 (3 H, d, J = 6.1); 0.87-0.79 (9 H, m)
H.2.6
Figure US08283367-20121009-C00115
5-Phenyl-pyridine-3-carboxamide, N-[(1S,2R)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-hydroxypropyl] M.p. 135-140° C. 1H-NMR (DMSO-d6): 9.06 (1 H, s); 9.02 (1 H, s); 9.00 (1 H, br. s); 8.66 (1 H, d, J = 8.5); 8.54 (1 H, br. s); 7.85 (2 H, d, J = 7.5); 7.57 (2 H, t, J = 7.5); 7.52-7.46 (1 H, m); 5.06 (1 H, d, J = 5.9); 4.58-4.52 (1 H, m); 4.12- 4.02 (2 H, m); 2.60 (1 H, br. t, J = 7.2); 2.28-2.18 (1 H, m); 2.08-1.98 (1 H, m); 1.84 (1 H, t, J = 5.4); 1.81-1.76 (1 H, m); 1.73-1.60 (2 H, m); 1.38-1.20 (3 H, m); 1.25 (3 H, s); 1.23 (3 H, s); 1.16 (3 H, d, J = 6.2); 0.86 (6 H, d, J = 6.3); 0.81 (3 H, s).
H.2.7
Figure US08283367-20121009-C00116
5-Phenyl-pyridine-2-carboxamide, N-[(1S,2R)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H-NMR (DMSO-d6): 9.08 (1 H, s); 9.01 (1 H, br. s); 8.68 (1 H, d, J = 8.5); 8.38 (1 H, d, J = 8.3); 8.19 (1 H, d, J = 8.0); 7.88 (2 H, d, J = 7.4); 7.62 (2 H, t, J = 7.3); 7.58-7.52 (1 H, m); 5.31 (1 H, br. d, J = 4.3); 4.55-4.49 (1 H, m); 4.20-4.12 (2 H, m); 2.68 (1 H, br. t, J = 7.2); 2.32-2.22 (1 H, m); 2.13-2.04 (1 H, m); 1.95-1.82 (2 H, m); 1.79-1.65 (2 H, m); 1.45-1.27 (3 H, m); 1.30 (3 H, s); 1.28 (3 H, s); 1.17 (3 H, d, J = 6.0); 0.90 (6 H, d, J = 6.4), 0.86 (3 H, s).
H.2.8
Figure US08283367-20121009-C00117
4-Phenyl-pyridine-2-carboxamide, N-[(1S,2R)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H NMR (DMSO-d6): 8.94 (1 H, br. s); 8.76 (1 H, d, J = 5.1); 8.67 (1 H, d, J = 8.5); 8.31 (1 H, s); 7.99 (1 H, d, J = 5.1); 7.88 (2 H, d, J = 7.4); 7.60-7.50 (3 H, m); 5.25 (1 H, d, J = 4.8); 4.47 (1 H, dd, J = 8.4, 4.1); 4.14-4.06 (2 H, m); 2.62 (1 H, br. t, J = 8.0); 2.26-2.16 (1 H, m); 2.06-1.98 (1 H, m); 1.84 (1 H, t, J = 5.5); 1.83-1.76 (1 H, m); 1.73-1.59 (2 H, m); 1.40- 1.20 (3 H, m); 1.24 (3 H, s); 1.22 (3 H, s); 1.11 (3 H, d, J = 6.2); 0.84 (6 H, d, J = 6.5); 0.80 (3 H, s).
H.2.9
Figure US08283367-20121009-C00118
Isoquinoline-1-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H-NMR (DMSO-d6): 9.14 (1 H, d, J = 8.6); 8.96 (1 H, s); 8.75 (1 H, d, J = 8.2); 8.60 (1 H, d, J = 5.5); 8.11-8.05 (2 H, m); 7.84 (1 H, t, J = 7.5); 7.73 (1 H, t, J = 7.5); 5.22 (1 H, d, J = 4.9); 4.51-4.46 (1 H, m); 4.15-4.06 (2 H, m); 2.64-2.57 (1 H, m); 2.24-2.15 (1 H, m); 2.06-1.97 (1 H, m); 1.83 (1 H, t, J = 5.0); 1.82-1.75 (1 H, m); 1.74-1.55 (2 H, m); 1.37-1.20 (3 H, m); 1.24 (3 H, s); 1.21 (3 H, s); 1.16 (3 H, d, J = 6.0), 0.85-0.78 (9 H, m).
H.2.10
Figure US08283367-20121009-C00119
Isoquinoline-3-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H NMR (DMSO-d6): 9.43 (1 H, s); 9.18 (1 H, s); 8.96 (1 H, s); 8.78 (1 H, d, J = 8.3); 8.28-8.20 (2 H, m); 7.90- 7.80 (2 H, m); 5.26 (1 H, d, J = 4.6); 4.52-4.48 (1 H, m); 4.11 (2 H, d, J = 7.0); 2.66-2.59 (1 H, m); 2.25-2.15 (1 H, m); 2.07-1.98 (1 H, m); 1.80-1.70 (2 H, m); 1.65-1.60 (2 H, m); 1.40-1.30 (3 H, m); 1.25 (3 H, s); 1.22 (3 H, s); 1.13 (3 H, d, J = 6.1); 0.87-0.79 (9 H, m)
H.2.11
Figure US08283367-20121009-C00120
Quinoline-3-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H-NMR (DMSO-d6): 9.31 (1 H, s); 9.00 (1 H, s); 8.91 (1 H, s); 8.61 (1 H, d, J = 8.3); 8.15-8.08 (2 H, m); 7.89 (1 H, t, J = 7.6); 7.72 (1 H, t, J = 7.6); 5.08 (1 H, d, J = 5.8); 4.58-4.52 (1 H, m); 4.12-4.00 (2 H, m); 2.59 (1 H, br. t); 2.25-2.15 (1 H, m); 2.04-1.97 (1 H, m); 1.86-1.80 (1 H, m); 1.80-1.74 (1 H, m); 1.73-1.58 (2 H, m); 1.38-1.20 (3 H, m); 1.24 (3 H, s); 1.21 (3 H, s); 1.16 (3 H, d, J = 6.4); 0.84 (6 H, d, J = 6.4); 0.80 (3 H, s).
H.1.12
Figure US08283367-20121009-C00121
5-(Thiophene-2-yl)pyridine-3-carboxamide, N- [(1S,2R)-1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol- 2-yl]-3-methylbutyl]amino]carbonyl]-2- hydroxypropyl] 1H-NMR (DMSO-d6): 9.06 (1 H, d, J = 1.9); 9.02 (1 H, s); 8.94 (1 H, d, J = 1.9); 8.66 (1 H, d, J = 8.2); 8.43 (1 H, t, J = 1.8); 7.77-7.70 (2 H, m); 7.26-7.21 (1 H, m); 5.06 (1 H, d, J = 5.8); 4.53-4.48 (1 H, m); 4.10- 4.09 (2 H, m); 2.60-2.54 (1 H, m); 2.24-2.14 (1 H, m); 2.05-1.96 (1 H, m); 1.82 (1 H, t, J = 5.4); 1.80-1.74 (1 H, m); 1.73-1.58 (2 H, m); 1.36-1.16 (3 H, m); 1.23 (3 H, s); 1.21 (3 H, s); 1.13 (3 H, d, J = 6.2); 0.86- 0.82 (6 H, m); 0.79 (3 H, s).
H.2.13
Figure US08283367-20121009-C00122
5-Phenyl-2H-pyrazole-3-carboxamide, N-[(1S,2R)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H-NMR (DMSO-d6): 13.6 (1 H, br.s); 9.03 (1 H, s); 7.91 (1 H, br. s); 7.88 (2 H, d, J = 7.7); 7.57-7.50 (2 H, m); 7.47-7.41 (1 H, m); 7.25 (1 H, br. s); 5.23 (1 H, br.s); 4.55-4.49 (1 H, m); 4.16 (1 H, d, J = 8.3); 4.13-4.06 (1 H, m); 2.69-2.62 (1 H, m); 2.31-2.22 (1 H, m); 2.13-2.05 (1 H, m); 1.90 (1 H, t, J = 5.5); 1.87-1.82 (1 H, m); 1.79-1.66 (2 H, m); 1.44-1.25 (3 H, m); 1.31 (3 H, s); 1.28 (3 H, s); 1.16 (3 H, d, J = 6.2); 0.91 (6 H, d, J = 6.5); 0.87 (3 H, s).
H.2.14
Figure US08283367-20121009-C00123
1H-Indole-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H NMR (DMSO-d6): 11.68 (1 H, s); 9.00 (1 H, s); 8.17 (1 H, d, J = 8.5); 7.65 (1 H, d, J = 8.0); 7.44 (1 H, d, J = 8.2); 7.25 (1 H, s); 7.20 (1 H, t, J = 7.5); 7.05 (1 H, t, J = 7.5); 5.08 (1 H, d, J = 5.9); 4.56-4.50 (1 H, m); 4.11-4.00 (2 H, m); 3.41-3.38 (1 H, m); 2.62-2.55 (1 H, m); 2.25-2.15 (1 H, m); 2.06-1.96 (1 H, m); 1.86-1.75 (2 H, m); 1.75-1.59 (2 H, m); 1.36-1.26 (3 H, m); 1.24 (3 H, s); 1.22 (3 H, s); 1.15- 1.07 (3 H, m); 0.87-0.79 (9 H, m).
H.2.15
Figure US08283367-20121009-C00124
6-Phenyl-pyrimidine-4-carboxamide, N-[(1S,2R)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H-NMR (DMSO-d6): 9.43 (1 H, s); 8.88 (1 H, br. s); 8.70 (1 H, d, J = 8.5); 8.52 (1 H, s); 8.33-8.28 (2 H, m); 7.64-7.56 (3 H, m); 5.27 (1 H, d, J = 4.8); 4.46 (1 H, dd, J = 7.9, 4.2); 4.16-4.07 (2 H, m); 2.69-2.62 (1 H, m); 2.26-2.17 (1 H, m); 2.08-1.98 (1 H, m); 1.84 (1 H, t, J = 5.2); 1.82-1.76 (1 H, m); 1.71-1.59 (2 H, m); 1.39- 1.20 (3 H, m); 1.24 (3 H, s); 1.22 (3 H, s); 1.12 (3 H, d, J = 6.7); 0.84 (6 H, d, J = 6.4); 0.80 (3 H, s).
H.2.16
Figure US08283367-20121009-C00125
5-Methyl-1-phenyl-1H-pyrazole-4-carboxamide, N- [(1S,2R)-1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol- 2-yl]-3-methylbutyl]amino]carbonyl]-2- hydroxypropyl] 1H-NMR (DMSO-d6): 9.20 (1 H, br. s); 8.30 (1 H, s); 7.91 (1 H, d, J = 8.5); 7.66-7.52 (5 H, m); 5.14 (1 H, d, J = 6.1); 4.61-4.58 (1 H, m); 4.14-4.04 (2 H, m); 2.62- 2.55 (4 H, m); 2.30-2.20 (1 H, m); 2.12-2.03 (1 H, m); 1.91-1.82 (2 H, m); 1.80-1.64 (2 H, m); 1.41 (1 H, d, J = 10.0); 1.38-1.25 (2 H, m); 1.31 (3 H, s); 1.28 (3 H, s); 1.17 (3 H, d, J = 6.3); 0.94-0.89 (6 H, m); 0.87 (3 H, s).
H.2.17
Figure US08283367-20121009-C00126
2-Phenyl-thiazole-4-carboxamide, N-[(1S,2R)-1- [[[(1R)-1-(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H NMR (DMSO-d6): 8.99-8.66 (1 H, m); 8.39 (1 H, s); 8.15 (1 H, d, J = 8.4); 8.06-8.01 (2 H, m); 7.58- 7.52 (3 H, m); 5.24 (1 H, d, J = 8.4); 4.48-4.42 (1 H, m); 4.14-4.06 (2 H, m); 2.65-2.58 (1 H, m); 2.26-2.16 (1 H, m); 2.08-2.00 (1 H, m); 1.85 (1 H, t, J = 5.4); 1.80-1.75 (1 H, m); 1.73-1.59 (2 H, m); 1.41-1.30 (2 H, m); 1.25 (3 H, s); 1.22 (3 H, s); 1.20-1.15 (1 H, m); 1.12 (3 H, d, J = 5.3); 0.85 (6 H, d, J = 6.5); 0.80 (3 H, s).
H.2.18
Figure US08283367-20121009-C00127
6-(Thiophene-2-yl)pyridine-2-carboxamide, N- [(1S,2R)-1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol- 2-yl]-3-methylbutyl]amino]carbonyl]-2- hydroxypropyl] 1H NMR (DMSO-d6): 8.93 (1 H, br. s); 8.58 (1 H, d, J = 8.2); 8.13 (1 H, d AB, J = 7.9); 8.04 (1 H, d AB, J = 7.9); 7.95-7.88 (2 H, m); 7.73 (1 H, d, J = 4.7); 7.22 (1 H, t, J = 4.2); 5.26 (1 H, d, J = 4.4); 4.44 (1 H, dd, J = 8.0, 4.1); 4.16-4.05 (2 H, m); 2.61 (1 H, br. t, J = 6.6); 2.25-2.15 (1 H, m); 2.08-1.98 (1 H, m); 1.84 (1 H, t, J = 5.2); 1.82-1.74 (1 H, m); 1.74-1.59 (2 H, m); 1.41-1.20 (3 H, m); 1.24 (3 H, s); 1.22 (3 H, s); 1.12 (3 H, d, J = 6.5); 0.84 (6 H, d, J = 6.4); 0.80 (3 H, s).
H.2.19
Figure US08283367-20121009-C00128
6-Butyl-pyridine-2-carboxamide, N-[(1S,2R)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-hydroxypropyl]
H.2.20
Figure US08283367-20121009-C00129
Pyridine-1-oxo-2-carboxamide, N-[(1S,2R)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] 1H-NMR (DMSO-d6): 11.54 (1 H, d, J = 7.6, NH); 8.96 (1 H, br. s); 8.46 (1 H, d, J = 6.3); 8.22 (1 H, d, J = 7.6); 7.68-7.57 (2 H, m); 5.17 (1 H, d, J = 4.2); 4.42-4.47 (1 H, m); 4.13-4.6 (2 H, m); 2.57 (1 H, br. t, J = 7.3); 2.25-2.16 (1 H, m); 2.07-1.98 (1 H, m); 1.84 (1 H, t, J = 5.6); 1.82-1.77 (1 H, m); 1.73-1.60 (2 H, m); 1.38-1.20 (3 H, m); 1.25 (3 H, s); 1.23 (3 H, s); 1.12 (3 H, d, J = 6.2); 0.85-0.80 (9 H, m).
H.2.21
Figure US08283367-20121009-C00130
Pyridine-1-oxo-3-carboxamide, N-[(1S,2R)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl]
Example J.1 Preparation of 2-Pyrazinecarboxamide, N-[(1S)-1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a, 5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2-[(4-methylbenzoylamide)ethyl]-
Figure US08283367-20121009-C00131
2-Pyrazinecarboxylic acid (163 mg, 1.31 mmol, 1.1 eq.) was dissolved in dry DMF (6 ml). TBTU (420 mg, 1.11 mmol, 1.1 eq.) was added to the solution at r.t. under nitrogen. The resulting mixture was stirred for 10 min and then cooled to 0-5° C. NMM (0.4 ml, 3.57 mmol, 3 eq.) and (2S)-2-amino-3-[(4-methylbenzoyl)amino]propanamide, N-[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]-, hydrochloride salt (600 mg, 1.19 mmol, 1 eq.) of Example C.4 were then added and the resulting mixture was stirred at r.t. for 5 h. The solution was poured into water (50 ml) and citric acid 2% (30 ml), then the aqueous suspension was extracted with ethyl acetate (40 ml). The organic solvent was washed with a solution of sodium bicarbonate 2% (30 ml) and NaCl 2% (50 ml). The organic phase was dried over anhydrous sodium sulphate, filtered, and evaporated. The resulting crude was purified by silica gel chromatography (eluent: ethyl acetate 100%). The collected fractions were evaporated under reduced pressure to a foam that was resuspended in diethyl ether (20 ml) for 30 min. The suspension was filtered and dried to give 330 mg of white foam. Yield 73%.
1H NMR (DMSO-d6): δ 9.14 (1H, s); 8.95 (1H, d); 8.9 (1H, d); 8.8 (1H, d); 8.78 (1H, d); 8.4 (1H, m); 7.7 (2H, d); 7.2 (2H, d); 4.8 (1H, q); 4.05 (1H, d); 3.65 (2H, m); 2.7 (1H, m); 2.35 (3H, s); 2.2 (1H, m); 2.05 (1H, m); 1.80 (2H, m); 1.60 (2H, m); 1.3-1.0 (10H) m); 0.8 (9H, m).
Example J.2 Preparation of Further Compounds of the Invention
Further compounds of the invention are listed below which can be made according to procedures analogous to those described for Example J.1 using the appropriate carboxylic acid such as described in Example M.1. Those compounds listed below which are characterized by NMR data were actually prepared.
Ex # Structure Chemical Name and Analytical Data
J.2.1
Figure US08283367-20121009-C00132
6-phenyl-2-pyridinocarboxamide, N-[(1S)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]- 1H NMR (DMSO-d6): 9.14 (1 H, d); 8.90 (1 H, d); 8.5 (1 H, t); 8.35 (2 H, d); 8.18 (1 H, d); 8.1 (1 H, t); 7.95 (1 H, d); 7.75 (2 H, d); 7.55 (3 H, m); 7.2 (2 H, d); 4.75 (1 H, m); 4.15 (1 H, d); 3.7 (2 H, m); 2.30 (3 H, s); 2.2 (1 H, m); 2.05 (1 H, m); 1.80 (2 H, m); 1.60 (2 H, m); 1.3-1.0 (6 H, m); 0.8 (6 H, m).
J.2.2
Figure US08283367-20121009-C00133
5-butyl-2-pyridinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.3
Figure US08283367-20121009-C00134
2-pyridinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]- 1H-NMR (DMSO-d6): 8.95 (1 H, d, J = 8.1); 8.89 (1 H, br. d, J = 3.3); 8.73 (1 H, d, J = 4.5); 8.48 (1 H, t, J = 5.7); 8.10-8.01 (2 H, m); 7.74 (2 H, d, J = 7.9); 7.68 (1 H, t, J = 5.3); 7.29 (2 H, d, J = 7.9); 4.87 (1 H, dd, J = 7.1, 13.3); 4.18 (1 H, d, J = 8.3); 3.78-3.64 (2 H, m); 2.78- 2.70 (1 H, m); 2.39 (3 H, s); 2.30-2.20 (1 H, m); 2.13- 2.04 (1 H, m); 1.92-1.81 (2 H, m); 1.72-1.63 (2 H, m); 1.46-1.29 (3 H, m); 1.29 (3 H, s); 1.27 (3 H, s); 0.89- 0.83 (9 H, m).
J.2.4
Figure US08283367-20121009-C00135
3-pyridinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]- 1H-NMR (DMSO-d6): 9.07 (1 H, s); 8.94 (2 H, m); 8.78 (1 H, d, J = 4.8); 8.55 (1 H, t, J = 4.8); 8.24 (1 H, d, J = 7.6); 7.77 (2 H, d, J = 7.9); 7.59 (1 H, dd, J = 7.8, 5.0); 7.31 (2 H, d, J = 7.9); 4.86 (1 H, dd, J = 7.7, 13.6); 4.17 (1 H, d, J = 8.0); 3.80-3.67 (2 H, m); 2.74-2.66 (1 H, m); 2.40 (3 H, s); 2.30-2.22 (1 H, m); 2.12-2.06 (1 H, m); 1.89 (1 H, t, J = 5.4); 1.87-1.81 (1 H, m); 1.75-1.65 (2 H, m); 1.45-1.29 (3 H, m); 1.30 (3 H, s); 1.28 (3 H, s); 0.89-0.83 (9 H, m).
J.2.5
Figure US08283367-20121009-C00136
Quinoline-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]- 1H NMR (DMSO-d6): 9.07 (1 H, d, J = 8.0); 8.92 (1 H, br. s); 8.58 (1 H, d, J = 8.6); 8.49 (1 H, t, J = 5.4); 8.17- 8.06 (3 H, m); 7.91 (1 H, t, J = 7.6); 7.77-7.69 (3 H, m); 7.23 (2 H, d, J = 7.8); 4.84 (1 H, dd, J = 13.9, 7.1); 4.14 (1 H, d, J = 8.3); 3.78-3.64 (2 H, m); 2.72-2.65 (1 H, m); 1.32 (3 H, s); 2.24-2.15 (1 H, m); 2.08-1.99 (1 H, m); 1.86-1.76 (2 H, m); 1.68-1.60 (2 H, m); 1.43-1.22 (3 H, m); 1.25 (3 H, s); 1.21 (3 H, s); 0.83-0.76 (9 H, m).
J.2.6
Figure US08283367-20121009-C00137
Quinoxaline-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]- 1H-NMR (DMSO-d6): 9.48 (1 H, s); 8.13 (1 H, d, J = 8.0); 8.88 (1 H, br. d, J = 3.5); 8.50 (1 H, t, J = 5.7); 8.26-8.20 (2 H, m); 8.06-8.01 (2 H, m); 7.73 (2 H, d, J = 7.9); 7.25 (2 H, d, J = 7.9); 4.88 (1 H, q, J = 6.8); 4.16 (1 H, d, J = 8.1); 3.76 (2 H, t, J = 6.0); 2.75-2.67 (1 H, m); 2.34 (3 H, s); 2.26-2.17 (1 H, m); 2.09-2.01 (1 H, m); 1.86 (1 H, t, J = 5.4); 1.83-1.78 (1 H, m); 1.70-1.62 (2 H, m); 1.43-1.25 (3 H, m); 1.27 (3 H, s); 1.23 (3 H, s); 0.85-0.79 (9 H, m).
J.2.7
Figure US08283367-20121009-C00138
6-phenyl-2-pyrazinocarboxamide, N-[(1S)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]- 1H NMR (DMSO-d6): 9.5 (1 H, s); 9.22 (1 H, d); 9.1 (1 H, s); 8.82 (1 H, bs); 8.6 (1 H, m); 8.4 (2 H, d); 7.75 (2 H, d); 7.6 (3 H, m); 7.2 (2 H, d); 4.75 (1 H, m); 4.1 (1 H, d), 3.75 (2 H, m); 2.35 (3 H, s); 2.7 (1 H, m); 2.2 (1 H, m); 2.05 (1 H, m); 1.80 (2 H, m); 1.60 (2 H, m); 1.3-1.0 (10 H, m); 0.8 (9 H, m).
J.2.8
Figure US08283367-20121009-C00139
5-phenyl-2-pyrazinocarboxamide, N-[(1S)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]- 1H NMR (DMSO-d6): 9.35 (1 H, s); 9.22 (1 H, s); 8.95 (1 H, d); 8.85 (1 H, d); 8.45 (1 H, t); 8.25 (1 H, d); 7.7 (2 H, d); 7.6 (3 H, m); 7.25 (2 H, d); 4.85 (1 H, q); 4.1 (1 H, d); 3.7 (2 H, m); 2.7 (1 H, m); 2.35 (3 H, s); 2.2 (1 H, m); 2.05 (1 H, m); 1.80 (2 H, m); 1.60 (2 H, m); 1.3-1.0 (9 H, m); 0.8 (9 H, m).
J.2.9
Figure US08283367-20121009-C00140
5-Phenyl-pyridine-3-carboxamide, N-[(1S)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.10
Figure US08283367-20121009-C00141
5-Phenyl-pyridine-2-carboxamide N-[(1S)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-,
J.2.11
Figure US08283367-20121009-C00142
4-Phenyl-pyridine-2-carboxamide, N-[(1S)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.12
Figure US08283367-20121009-C00143
Isoquinoline-1-carboxamide N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-,
J.2.13
Figure US08283367-20121009-C00144
Isoquinoline-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.14
Figure US08283367-20121009-C00145
Quinoline-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.15
Figure US08283367-20121009-C00146
5-(Thiophene-2-yl)-pyridine-3-carboxamide, N-[(1S)- 1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.16
Figure US08283367-20121009-C00147
5-Phenyl-2H-pyrazole-3-carboxamide, N-[(1S)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.17
Figure US08283367-20121009-C00148
1H-Indole-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.18
Figure US08283367-20121009-C00149
6-Phenyl-pyrimidine-4-carboxamide, N-[(1S)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.19
Figure US08283367-20121009-C00150
5-Methyl-1-phenyl-1H-pyrazole-4-carboxamide N- [(1S)-1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro- 3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol- 2-yl]-3-methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.20
Figure US08283367-20121009-C00151
2-Phenyl-thiazole-4-carboxamide, N-[(1S)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.21
Figure US08283367-20121009-C00152
6-(Thiophene-2-yl)pyridine-2-carboxamide, N-[(1S)- 1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]- 1H NMR (DMSO-d6): 8.90-8.86 (1 H, m); 8.84 (1 H, d, J = 7.7); 8.47 (1 H, t, J = 5.8); 8.07-7.98 (3 H, m); 7.87 (1 H, d, J = 7.3); 7.77-7.70 (3 H, m); 7.27-7.20 (3 H, m); 4.77 (1 H, m); 4.15 (1 H, d, J = 7.7); 3.74- 3.64 (2 H, m); 2.72-2.65 (1 H, m); 2.33 (3 H, s); 2.24- 2.15 (1 H, m); 2.09-2.00 (1 H, m); 1.84 (1 H, t, J = 5.3); 1.80-1.75 (1 H, m); 1.68-1.59 (2 H, m); 1.42-1.32 (2 H, m); 1.29-1.23 (1 H, m); 1.26 (3 H, s); 1.21 (3 H, s); 0.83-0.77 (9 H, m).
J.2.22
Figure US08283367-20121009-C00153
6-Butyl-pyridine-2-carboxamide, N-[(1S)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.23
Figure US08283367-20121009-C00154
6-morpholino-3-pyridinocarbonylamino, N-[(1S)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]- 3-methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]- 1H NMR (DMSO-d6): 8.9 (1 H, d); 8.90 (1 H, d); 8.65 (1 H, d); 8.45 (2 H, m); 8.0 (1 H, d); 7.75 (2 H, d); 7.3 (2 H, d); 6.95 (1 H, d); 4.8 (1 H, q); 4.15 (1 H, d); 3.75 (4 H, m); 3.70 (2 H, m); 3.6 (4 H, m); 2.6 (1 H, m); 2.35 (3 H, s); 2.2 (1 H, m); 1.85 (2 H, m); 1.65 (2 H, m); 1.3- 1.0 (8 H, m); 0.85 (9 H, m).
J.2.24
Figure US08283367-20121009-C00155
Pyridine-1-oxo-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
J.2.25
Figure US08283367-20121009-C00156
Pyridine-1-oxo-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-[(4- methylbenzoylamide)ethyl]-
Example K.1 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(6-phenyl-pyrazine-2-carbonyl)amino]-1-oxobutyl]amino]-3-methylbutyl]
Figure US08283367-20121009-C00157
A mixture of 6-phenyl-pyrazine-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2-hydroxypropyl] of Example H.1 (235 mg, 0.428 mmol) and 2-methylpropylboronic acid (131 mg, 1.28 mmol) was dissolved in methanol (3 ml). Then 2 N aqueous HCl (0.6 ml) and hexane (3 ml) were added. The resulting heterogeneous mixture was vigorously stirred at room temperature for 16 hours. The layers were then separated and the methanolic phase was concentrated to dryness. The residue was purified by elution on a SPE-SI normal phase cartridge (20 g SiO2) using first ethyl acetate, then methanol. The product was then dissolved in ethyl acetate containing 4% methanol (100 ml) and washed with a 10% solution of NaHCO3. The phases were separated and the organic layer was dried over sodium sulfate and concentrated to dryness. The product was obtained as a white solid (120 mg, 70% yield).
1H NMR (MeOH-d4): δ 9.40 (1H, s); 9.22 (1H, s); 8.29-8.25 (2H, m); 7.61-7.58 (3H, m); 4.80 (1H, d, J=6.9); 4.47-4.41 (1H, m); 2.78 (1H, t, J=7.5); 1.71-1.61 (1H, m); 1.37 (2H, t, J=7.3); 1.31 (3H, d, J=6.4); 0.93 (3H, s); 0.91 (3H, s).
Example K.2 Preparation of Further Compounds of the Invention
Further compounds of the invention are listed below which can be made according to procedures analogous to those described for Example K.1 using the appropriate boronic ester. Those compounds listed below which are characterized by NMR data were actually prepared.
Ex # Structure Chemical Name and Analytical Data
K.2.1
Figure US08283367-20121009-C00158
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(pyridine-2-carbonyl)amino]-1-oxobutyl]amino]-3- methylbutyl]
K.2.2
Figure US08283367-20121009-C00159
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(pyridine-3-carbonyl)amino]-1-oxobutyl]amino]-3- methylbutyl]
K.2.3
Figure US08283367-20121009-C00160
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(quinoline-2-carbonyl)amino]-1-oxobutyl]amino]-3- methylbutyl] 1H NMR (MeOH-d4): 8.41 (1 H, m); 8.15-8.09 (2 H, m); 7.93 (1 H, m); 7.77 (1 H, t, J = 7.7); 7.62 (1 H, t, J = 7.5); 4.69 (1 H, d, J = 3.2); 4.36-4.29 (1 H, m); 2.66 (1 H, t, J = 7.6); 1.59-1.50 (1 H, m); 1.25, t, J = 7.3); 1.20 (3 H, d, J = 6.4); 0.81 (3 H, s); 0.79 (3 H, s)
K.2.4
Figure US08283367-20121009-C00161
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(quinoxaline-2-carbonyl)amino]-1-oxobutyl]amino]-3- methylbutyl] 1H NMR (MeOH-d4): 9.53 (1 H, s); 8.30-8.25 (1 H, m); 8.22-8.17 (1 H, m); 8.02-7.94 (2 H, m); 4.80 (1 H, d, J = 3.1); 4.46-4.39 (1 H, m); 2.76 (1 H, t, J = 7.6); 1.69-1.58 (1 H, m); 1.35 (2 H, t, J = 7.3); 1.30 (3 H, d, J = 6.4); 0.89 (6 H, d, J = 6.5).
K.2.5
Figure US08283367-20121009-C00162
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(5- phenyl-pyrazine-2-carbonyl)amino]-1-oxobutyl]amino]- 3-methylbutyl] 1H NMR (MeOH-d4): 9.29 (1 H, s); 9.23 (1 H, s); 8.23- 8.18 (2 H, m); 7.60-7.54 (3 H, m); 4.75 (1 H, d, J = 6.9); 4.44-4.35 (1 H, m); 2.76 (1 H, t, J = 6.7); 1.72-1.58 (1 H, m); 1.36 (2 H, t, J = 7.3); 1.28 (3 H, d, J = 6.4); 0.91 (6 H, d, J = 6.5).
K.2.6
Figure US08283367-20121009-C00163
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(5- phenyl-pyridine-3-carbonyl)amino]-1-oxobutyl]amino]- 3-methylbutyl] 1H NMR (MeOH-d4): 9.04 (1 H, s); 9.01 (1 H, s); 8.58 (1 H, s); 7.78 (2 H, d, J = 7.7); 7.59-7.53 (2 H, m); 7.52- 7.46 (1 H, m); 4.82 (1 H, d, J = 4.5); 4.38-4.31 (1 H, m); 2.80 (1 H, t, J = 7.6); 1.73-1.62 (1 H, m); 1.38 (2 H, t, J = 7.3); 1.32 (3 H, d, J = 6.3); 0.94 (6 H, d, J = 6.5).
K.2.7
Figure US08283367-20121009-C00164
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(5- phenyl-pyridine-2-carbonyl)amino]-1-oxobutyl]amino]- 3-methylbutyl] M.p. 190-195° C. 1H NMR (MeOH-d4): 8.98 (1 H, s); 8.27-8.16 (2 H, m); 7.75 (2 H, d, J = 7.3); 7.56-7.51 (2 H, m); 7.50-7.44 (1 H, m); 4.74 (1 H, br. d, J = 3.0); 4.42-4.34 (1 H, m); 2.75 (1 H, t, J = 7.5); 1.70-1.59 (1 H, m); 1.35 (2 H, t, J = 7.3); 1.27 (3 H, d, J = 6.3); 0.93-0.89 (6 H, m).
K.2.8
Figure US08283367-20121009-C00165
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(4- phenyl-pyridine-2-carbonyl)amino]-1-oxobutyl]amino]- 3-methylbutyl] 1H NMR (MeOH-d4): 8.75 (1 H, d, J = 4.8); 8.41 (1 H, s); 7.91 (1 H, d, J = 4.9); 7.82 (2 H, d, J = 7.5); 7.60-7.47 (3 H, m); 4.77 (1 H, br. s); 4.45-4.36 (1 H, m); 2.77 (1 H, t, J = 7.5); 1.72-1.61 (1 H, m); 1.37 (2 H, t, J = 7.3); 1.29 (3 H, d, J = 6.3); 0.92 (6 H, d, J = 6.3).
K.2.9
Figure US08283367-20121009-C00166
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(isoquinoline-1-carbonyl)amino]-1-oxobutyl]amino]-3- methylbutyl] 1H NMR (MeOH-d4): 9.02 (1 H, d, J = 8.7); 8.47 (1 H, d, J = 5.5); 7.92-7.88 (2 H, m); 7.71 (1 H, t, J = 7.5); 7.62 (1 H, t, J = 7.7); 4.71 (1 H, d, J = 3.1); 4.33-4.25 (1 H, m); 2.65 (1 H, t, J = 7.5); 1.61-1.49 (1 H, m); 1.26 (2 H, t, J = 7.7); 1.21 (3 H, d, J = 6.3); 0.80 (6 H, d, J = 6.5).
K.2.10
Figure US08283367-20121009-C00167
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(isoquinoline-3-carbonyl)amino]-1-oxobutyl]amino]-3- methylbutyl] 1H NMR (MeOH-d4): 9.35 (1 H, s); 8.57 (1 H, s); 8.20 (1 H, d, J = 8.0); 8.11 (1 H, d, J = 8.0); 7.91-7.79 (2 H, m); 4.79 (1 H, s); 4.45-4.35 (1 H, m); 2.75 (1 H, t, J = 7.3); 1.55-1.51 (1 H, m); 1.35 (2 H, t, J = 7.3); 1.29 (3 H, d, J = 6.4); 0.91 (3 H, s); 0.91 (3 H, s)
K.2.11
Figure US08283367-20121009-C00168
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(quinoline-3-carbonyl)amino]-1-oxobutyl]amino]-3- methylbutyl] 1H NMR (MeOH-d4): 9.20 (1 H, s); 8.82 (1 H, s); 8.01 (2 H, t, J = 7.5); 7.82 (1 H, t, J = 7.7); 7.63 (1 H, t, J = 7.6); 4.72 (1 H, br. s); 4.28-4.24 (1 H, m); 2.68 (1 H, t, J = 7.6); 1.62-1.50 (1 H, m); 1.27 (2 H, t, J = 7.3); 1.22 (3 H, d, J = 6.3); 0.82 (6 H, d, J = 6.5).
K.2.12
Figure US08283367-20121009-C00169
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(5- (thiophene-2-yl)pyridine-3-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] 1H NMR (MeOH-d4): 9.03 (1 H, s); 8.96 (1 H, s); 8.55 (1 H, s); 7.66 (1 H, d, J = 3.2); 7.59 (1 H, d, J = 4.8); 7.25- 7.20 (1 H, m); 4.81 (1 H, d, J = 4.6); 4.38-4.30 (1 H, m); 2.79 (1 H, t, J = 7.6); 1.73-1.62 (1 H, m); 1.38 (2 H, t, J = 7.3); 1.31 (3 H, d, J = 6.4); 0.94 (6 H, d, J = 6.5).
K.2.13
Figure US08283367-20121009-C00170
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(5- phenyl-2H-pyrazole-3-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] M.p. 232-236° C. 1H NMR (MeOH-d4): 7.75 (2 H, 1 br. d, J = 6.8); 7.52- 7.45 (2 H, m); 7.44-7.38 (1 H, m); 7.10 (1 H, br. s); 4.77 (1 H, br. d, J = 2.7); 4.39-4.32 (1 H, m); 2.77 (1 H, t, J = 7.4); 1.74-1.62 (1 H, m); 1.37 (2 H, t, J = 7.3); 1.29 (3 H, d, J = 6.4); 0.95-0.92 (6 H, m).
K.2.14
Figure US08283367-20121009-C00171
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(1 H- indole-2-carbonyl)amino]-1-oxobutyl]amino]-3- methylbutyl] 1H NMR (MeOH-d4): 7.64 (1 H, d, J = 8.0); 7.45 (1 H, d, J = 8.3); 7.24 (1 H, t, J = 8.0); 7.22 (1 H, bs); 7.08 (1 H, d, J = 7.5); 4.79 (1 H, d, J = 3.6); 4.37-4.30 (1 H, m); 2.75 (1 H, t, J = 7.3); 1.71-1.59 (1 H, m); 1.36 (2 H, t, J = 7.3); 1.28 (3 H, d, J = 6.3); 0.94-0.89 (6 H, m).
K.2.15
Figure US08283367-20121009-C00172
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(6- phenyl-pyrimidine-4-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] 1H NMR (MeOH-d4): 9.34 (1 H, d, J = 0.9); 8.56 (1 H, d, J = 0.9); 8.28-8.23 (2 H, m); 7.61-7.54 (3 H, m); 4.75 (1 H, d, J = 3.2); 4.43-4.35 (1 H, m); 2.76 (1 H, t, J = 7.6); 1.70- 1.59 (1 H, m); 1.35 (2 H, t, J = 7.3); 1.28 (3 H, d, J = 6.4); 0.91 (3 H, d, J = 6.5); 0.90 (3 H, d, J = 6.5).
K.2.16
Figure US08283367-20121009-C00173
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(5- Methyl-1-phenyl-1H-pyrazole-4-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] 1H NMR (MeOH-d4): 8.18 (1 H, s); 7.63-7.52 (3 H, m); 7.51-7.46 (2 H, m); 4.76 (1 H, d, J = 3.4); 4.36-4.29 (1 H, m); 2.77 (1 H, t, J = 7.6); 1.73-1.62 (1 H, m); 1.38 (2 H, t, J = 7.2); 1.30 (3 H, d, J = 6.4); 0.95 (6 H, d, J = 6.4).
K.2.17
Figure US08283367-20121009-C00174
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(2- phenyl-thiazole-4-carbonyl)amino]-1-oxobutyl]amino]- 3-methylbutyl] 1H NMR (MeOH-d4): 8.28 (1 H, s); 8.10-8.04 (2 H, m); 7.54-7.49 (3 H, m); 4.74 (1 H, d, J = 3.0); 4.44-4.35 (1 H, m); 2.75 (1 H, t, J = 7.6); 1.69-1.59 (1 H, m); 1.35 (2 H, t, J = 7.3); 1.29 (3 H, d, J = 6.4); 0.91 (6 H, d, J = 6.5).
K.2.18
Figure US08283367-20121009-C00175
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(6- (thiophene-2-yl)pyridine-2-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] 1H NMR (MeOH-d4): 8.06-7.95 (3 H, m); 7.84 (1 H, d, J = 3.7); 7.58 (1 H, d, J = 5.0); 7.21-7.17 (1 H, m); 4.71 (1 H, br. d, J = 3.0); 4.46-4.39 (1 H, m); 2.76 (1 H, t, J = 7.6); 1.71-1.61 (1 H, m); 1.37 (2 H, t, J = 7.3); 1.32 (3 H, d, J = 6.4); 0.92 (6 H, d, J = 6.6).
K.2.19
Figure US08283367-20121009-C00176
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(6-butyl- pyridine-2-carbonyl)amino]-1-oxobutyl]amino]-3- methylbutyl]
K.2.20
Figure US08283367-20121009-C00177
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(pyridine-1-oxo-2-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] 1H NMR (MeOH-d4): 8.45 (1 H, d, J = 6.0); 8.36 (1 H, dd, J = 7.6, 2.5); 7.74-7.64 (2 H, m); 4.73 (1 H, br. d, J = 3.0); 4.44-4.37 (1 H, m); 2.75 (1 H, t, J = 7.6); 1.72-1.61 (1 H, m); 1.40-1.33 (2 H, m); 1.31 (3 H, d, J = 6.3); 0.92 (6 H, d, J = 6.5).
K.2.21
Figure US08283367-20121009-C00178
Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(pyridine-1-oxo-3-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl]
Example L.1 Preparation of Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2-[(pyrazinocarbonylamino)]-1-oxopropyl]amino]-3-methylbutyl]
Figure US08283367-20121009-C00179
2-Pyrazinecarboxamide, N-[(1S)-1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2-[(4-methylbenzoylamide)ethyl]-, of Example J.1 (450 mg, 0.78 mmol, 1 eq.), was dissolved in methanol (4 ml), and n-hexane (4 ml). To this solution, isobutylboronic acid (120 mg, 1.17 mmol, 1.5 eq,) and HCl 4N 1,4-dioxane (0.3 ml, 1.17 mmol, 1.5 eq.) were added. The resulting biphasic mixture was stirred at room temperature for 48 h. The n-hexane was removed, and the resulting the methanolic solution was washed with n-hexane (2 ml) and evaporated under reduced pressure. The crude was redissolved in dichloromethane (250 ml) and washed with sodium bicarbonate 2%. The organic phase was dried over anhydrous sodium sulphate and evaporated under reduced pressure to give a foam. The foam was stirred in diethyl ether overnight, then was filtered, to give a white powder. Yield 44%, 150 mg.
Analytical data: M.p. 132° C.-135° C. A.E. calculated: C (57.16%), H (6.40%), N (15.87%). found C (56.56%), N (15.26%).
1H NMR (DMSO-d6): δ 9.25 (1H, s); 8.82 (1H, d); 8.72 (1H, d); 7.75 (2H, d); 7.25 (2H, d); 5.05 (1H, t); 3.95 (2H, m); 2.8 (1H, t); 2.4 (3H, s); 1.6 (1H, m); 1.35 (2H, m); 1.60 (2H, m); 1.3-1.0 (9H, m); 0.85 (6H, dd).
Example L.2 Preparation of Further Compounds of the Invention
Further compounds of the invention are listed below which can be made according to procedures analogous to those described for Example L.1 using the appropriate boronic ester. Those compounds listed below which are characterized by NMR data were actually prepared.
Ex # Structure Chemical Name and Analytical Data
L.2.1
Figure US08283367-20121009-C00180
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(6-phenyl-pyridine-2- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl] 1H NMR (DMSO-d6): 8.3 (2 H, d); 8.11 (1 H, m); 8.07 (2 H, d); 7.85 (2 H, d); 7.5 (2 H, d); 7.25 (2 H, d); 5.0 (1 H, t); 4.01-3.98 (2 H, m); 2.8 (1 H, t); 2.4 (3 H, s); 1.6 (1 H, m); 1.35 (2 H, m); 0.9 (6 H, dd).
L.2.2
Figure US08283367-20121009-C00181
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(5-butyl-pyridine-2- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl] 1H NMR (MeOH-d4): 8.52 (1 H, s); 7.97 (1 H, d, J = 8.0); 7.78 (1 H, d, J = 7.9); 7.71 (2 H,d,J = 7.8); 7.26 (2 H, d, J = 7.8); 5.01 (1 H, t, J = 6.2); 3.89 (2 H,d, J = 6.2); 2.80-2.70 (3 H, m); 2.38 (3 H, s); 1.70-1.53 (3 H, m); 1.44-1.27 (4 H, m); 0.96 (3 H, t, J = 7.3); 0.88 (3 H, d, J = 6.5); 0.84 (3 H, d, J = 6.5).
L.2.3
Figure US08283367-20121009-C00182
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(pyridine-2- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl] 1H NMR (MeOH-d4): 8.70 (1 H, br. s); 8.86 (1 H, d, J = 7.7); 8.0 1-7.94 (1 H, m); 7.73 (2 H, d, J = 7.2); 7.63- 7.55 (1 H, m); 7.28 (2 H, d, J = 7.7); 5.04 (1 H, t, J = 6.0); 3.91 (2 H, br. d, J = 5.8); 2.79 (1 H, t, J = 7.5); 2.40 (3 H, s); 1.66-1.54 (1 H, m); 1.34 (2 H, br. t, J = 7.1); 0.90 (3 H, d, J = 6.5); 0.86 (3 H, d, J = 6.6).
L.2.4
Figure US08283367-20121009-C00183
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(pyridine-3- carbonyl)amino]-1-oxopropyl]amino]-3-methyibutyl] M.p. 220-222° C. 1H NMR (MeOH-d4): 9.03 (1 H, s); 8.71 (1 H, d, J = 4.6); 8.28 (1 H, d, J = 8.0); 7.74 (2 H, d, J = 7.9); 7.57 (1 H, dd, J = 7.8, 5.0); 7.28 (2 H, d, J = 7.9); 4.98 (1 H, t, J = 6.4); 3.95 (1 H, dd, J = 13.8, 6.5, AB); 3.83 (1 H, dd, J = 13.8, 6.5, AB); 2.77 (1 H, t, J = 7.5); 2.39 (3 H, s); 1.63-1.52 (1 H, m); 1.37-1.26 (2 H, m); 0.88 (3 H, d, J = 6.5); 0.83 (3 H, d, J = 6.5).
L.2.5
Figure US08283367-20121009-C00184
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(quinoline-2- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl] M.p. 235-237° C. 1H NMR (MeOH-d4): 8.82 (1 H, br. t, J = 5.7); 8.47 (1 H, d, J = 8.5); 8.23 (1 H, d, J = 8.5); 8.15 (1 H, d, J = 8.5); 8.00 (1 H, d, J = 8.2); 7.86 (1 H, t, J = 7.6); 7.75 (2 H, d, J = 7.9); 7.70 (1 H, t, J = 7.6); 7.26 (2 H, d, J = 7.9); 5.08 (1 H, t, J = 6.2); 4.05-3.91 (2 H, m); 2.79 (1 H, t, J = 7.7); 2.38 (3 H, s); 1.67-1.56 (1 H, m); 1.34 (2 H, br. t, J = 7.3); 0.88 (3 H, d, J = 6.5); 0.85 (3 H, d, J = 6.5).
L.2.6
Figure US08283367-20121009-C00185
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(quinoxaline-2- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl] M.p. 160-1602° C. 1H NMR (MeOH-d4): 9.48 (1 H, s), 8.34-8-29 (1 H, m); 8.21-8.16 (1H, m); 8.01-7.94 (2 H, m); 7.74 (2 H, d, J = 7.9); 7.27 (2 H, d, J = 7.9); 5.08 (1 H, t, J = 6.1); 4.05-3.93 (2 H, m); 2.79 (1 H, t, J = 7.4); 2.38 (3 H, s); 1.66-1.55 (1 H, m); 1.35 (2 H, t, J = 7.3); 0.89 (3 H, d, J = 6.5); 0.85 (3 H, d, J = 6.5).
L.2.7
Figure US08283367-20121009-C00186
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(6-phenyl-2- pyrazinocarbonylamino)]-1-oxopropyl]amino]-3- methylbutyl 1H NMR (DMSO-d6): 9.35 (1 H, s); 9.11 (1 H, s); 8.4 (2 H, d); 7.80 (2 H, d); 7.6 (3 H, m); 7.25 (2 H, d); 5.0 (1 H, t); 4.01-3.98 (2 H, m); 2.8 (1 H, t); 2.35 (3 H, s); 1.6 (1 H, m); 1.35 (2 H, m); 0.9 (6 H, dd).
L.2.8
Figure US08283367-20121009-C00187
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(5-phenyl-2- pyrazinocarbonylamino)]-1-oxopropyl]amino]-3- methylbutyl 1H NMR (DMSO-d6): 9.25 (2 H, d); 8.2 (2 H, d); 7.75 (2 H, d); 7.65 (3 H, m); 7.25 (2 H, d); 5.05 (1 H, t); 3.95 (2 H, m); 2.8 (1 H, t); 2.35 (3 H, s); 1.6 (1 H, m); 1.35 (2 H, m); 0.9 (6 H, dd).
L.2.9
Figure US08283367-20121009-C00188
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(5-phenyl-pyridine-3- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
L.2.10
Figure US08283367-20121009-C00189
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(5-phenyl-pyridine-2- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
L.2.11
Figure US08283367-20121009-C00190
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(4-phenyl-pyridine-2- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
L.2.12
Figure US08283367-20121009-C00191
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(isoquinoline-1- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
L.2.13
Figure US08283367-20121009-C00192
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(isoquinoline-3- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
L.2.14
Figure US08283367-20121009-C00193
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(quinoline-3- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
L.2.15
Figure US08283367-20121009-C00194
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(5-(thiophene-2- yl)pyridine-3-carbonyl)amino]-1-oxopropyl]amino]- 3-methylbutyl]
L.2.16
Figure US08283367-20121009-C00195
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(5-phenyl-2H-pyrazole-3- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
L.2.17
Figure US08283367-20121009-C00196
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(1 H-indole-2- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
L.2.18
Figure US08283367-20121009-C00197
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(6-phenyl-pyrimidine-4- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
L.2.19
Figure US08283367-20121009-C00198
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(5-Methyl-1-phenyl-1H- pyrazole-4-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl]
L.2.20
Figure US08283367-20121009-C00199
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(2-phenyl-thiazole-4- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
L.2.21
Figure US08283367-20121009-C00200
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(6-(thiophene-2- yl)pyridine-2-carbonyl)]-1-oxopropyl]amino]-3- methylbutyl M.p. 158-160° C. 1H NMR (MeOH-d4): 8.01 (1 H, d, J = 3 .5); 7.98-7.92 (3 H, m); 7.81 (2 H, d, J = 7.9); 7.55 (1 H, d, J = 4.9); 7.28 (2 H, d, J = 7.9); 7.19 (1 H, t, J = 4.3); 5.02 (1 H, t, J = 5.2); 4.10-4.02 (1 H, m); 3.97-3.88 (1 H, m); 2.80 (1 H, t, J = 7.6); 2.40 (3 H, s); 1.67-1.56 (1 H, m); 1.36 (2 H, t, J = 7.3); 0.90 (3 H, d, J = 6.6); 0.87 (3 H, d, J = 6.6).
L.2.22
Figure US08283367-20121009-C00201
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(6-butyl-pyridine-2- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
L.2.23
Figure US08283367-20121009-C00202
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[(6-morpholino-3- pyridinocarbonylamino)]-1-oxopropyl]amino]-3- methylbutyl 1H NMR (DMSO-d6): 8.5 (1 H, s); 8.3 (1 H, d); 7.7 (2 H, d); 7.35 (1 H, d); 7.30 (2 H, d); 5.0 (1 H, t); 4.01- 3.75 (10 H, m); 2.8 (1 H, t); 2.4 (3 H, s); 1.6 (1 H, m); 1.35 (2 H, m); 0.9 (6 H, dd).
L.2.24
Figure US08283367-20121009-C00203
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[( pyridine-1-oxo-2- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
L.2.25
Figure US08283367-20121009-C00204
Boronic acid, [(1R)-1-[[(2 S)-3-[(4- methylbenzoyl)amino]-2-[( pyridine-1-oxo-3- carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]
Example M.1 Preparation/Source of Carboxylic Acids
Carboxylic acids suitable for preparing compounds of the invention according to, for example, the procedures of Examples H.1 and J.1 can be obtained commercially or prepared according to routine methods or known synthetic procedures. For example, 6-phenyl-pyrazine -2-carboxylic acid and 5-phenyl-pyrazine-2-carboxylic acid were prepared according to the procedure described in Litmanowitsch et al. CH458361. 6-Butyl-pyridine-2-carboxylic acid was prepared via a Reissert-Kaufman type reaction according the procedures described by Honma et al. in J. Med. Chem., 1984, 25, 125-128 or Nakashima et al., Ykugaku Zasshi, 1958, 78, 666-670 (Chem. Abstr. 1958, 18399). Compounds 6-phenyl-2-pyridinecarboxylic acid and 6-(thiophene-2-yl)pyridine-2-carboxylic acids were prepared according the procedure described by Gong et al. in Synlett, 2000, (6), 829-831.
Utility
Methods and Compositions
Compounds of the present invention can inhibit the activity of proteasome, leading to the inhibition or blocking of a variety of intracellular functions with which the proteasome is directly or indirectly associated. For example, proteasome inhibitors can modulate, such as induce, apoptosis in a cell. In some embodiments, the compounds herein can kill tumor cells by induction of apoptosis. Thus, the present compounds can be used to treat cancer, tumors or other proliferative disorders.
In further embodiments, inhibition of proteasome function by compounds of the invention can inhibit the activation or processing of transcription factor NF-κB. This protein plays a role in the regulation of genes involved in the immune and inflammatory responses as well as in cell viability. Inhibition of proteasome function can also inhibit the ubiquitination/proteolysis pathway. This pathway catalyzes, inter alia, selective degradation of highly abnormal proteins and short-lived regulatory proteins. In some embodiments, compounds of the invention can prevent the degradation of p53 which is typically degraded by the ubiquitin-dependent pathway. The ubiquitination/proteolysis pathway also is involved in the processing of internalized cellular or viral antigens into antigenic peptides that bind to MHC-I molecules. Thus, the compounds of the invention can be used to reduce the activity of the cytosolic ATP-ubiquitin-dependent proteolytic system in a number of cell types.
Accordingly, the usefulness of such compounds can include therapeutics, such as the treatment of various diseases or disorders associated with proteasome. The methods include administering a therapeutically effective amount of a compound of the invention, or composition thereof, to a mammal, such as a human having a disease or disorder associated with proteasome. The phrase “therapeutically effective amount” refers to an amount sufficient to prevent, alleviate, or ameliorate any phenomenon, such as a cause or symptom, known in the art to be associated with the disease or disorder.
Treatable diseases or disorders (abnormal physical conditions) can be associated with either normal or abnormal activities of proteasome, such as the regulation of apoptosis. Numerous diseases or disorders that are associated with proteasome, or that are desirably treated by induction of apoptosis, are known and include, for example, various cancers and tumors including those associated with skin, prostate, colorectal, pancreas, kidney, ovary, mammary, liver, tongue, lung, and smooth muscle tissues. Preferred tumors that can be treated with proteasome inhibitors include, but are not limited to hematological tumors, such as, for example, leukemias, lymphomas, non-Hodgkin lymphoma, myeloma, multiple myeloma, as well as solid tumors such as, for example, colorectal, mammary, prostate, lung, and pancreas tumors. In order to elicit therapeutic effects, the proteasome inhibitors can be administered to patients as single agents or in combination with one or more antitumor or anticancer agent and/or radiotherapy. Examples of other anti-tumor or anti-cancer agents which can be advantageously administered concomitantly with a proteasome inhibitor include but are not limited to, adriamycin, daunomycin, methotrexate, vincristin, 6-mercaptopurine, cytosine arabinoside, cyclophosphamide, 5-FU, hexamethylmelamine, carboplatin, cisplatin, idarubycin, paclitaxel, docetaxel, topotecan, irinotecam, gemcitabine, L-PAM, BCNU and VP-16. Methods for determining apoptosis in vitro are well known in the art and kits are available commercially. See for example the Apo-ONE™ Homogeneous Caspase-3/7 Assay from Promega Corporation, Madison Wis., USA (Technical Bulletin No. 295, revised 2/02, Promega Corporation).
Further diseases or disorders associated with the proteasome include accelerated or enhanced proteolysis that occurs in atrophying muscles, such as is often associated with activation of a nonlysomal ATP-requiring process involving ubiquitin. Accelerated or enhanced proteolysis can be the result of any of numerous causes including sepsis, burns, trauma, cancer, infection, neurodegenerative diseases such as muscular dystrophy, acidosis, or spinal/nerve injuries, corticosteroid use, fever, stress, and starvation. Compounds of the invention can be tested for inhibition of muscle wastage by any various procedures known in the art such as by measuring urinary excretion of modified amino acid 3-methylhistidine (see, e.g., Young, et al., Federation Proc., 1978, 37, 229).
Compounds of the present invention can be further used to treat or prevent diseases or disorders associated with activity of NF-κB including for example, human immunodeficiency virus (HIV) infection and inflammatory disorders resulting from, for example, transplantation rejection, arthritis, infection, inflammatory bowel disease, asthma, osteoporosis, osteoarthritis, psoriasis, restenosis, and autoimmune diseases. Accordingly, a process that prevents activation of NF-κB in patients suffering from such a disease would be therapeutically beneficial. Inhibition of the NF-κB activity can be measured by using a DNA binding assay such a described in Palombella, et al., Cell, 1994, 78, 773.
Those of ordinary skill in the art can readily identify individuals who are prone to or suspected of suffering from such diseases or disorders using standard diagnostic techniques.
Example A Assay for Chymotrypsin-like Activity of 20S Human Erythrocyte Proteasome (HEP)
Proteasome chymotrypsin-like activity of compounds of the invention can be assayed according to the following procedure.
In 96-well microtiter plates, 20S Human Erythrocyte Proteasome (HEP), purchased from Immatics Biotechnologies Inc., Tübingen, Germany can be plated at 0.2 μg/mL (about 0.6 nM catalytic sites) in 0.04% SDS 20 mM Tris buffer. A fluorimetric substrate Suc-LLVY-AMC (succinyl-Leu-Leu-Val-Tyr-7-amido-4-methylcoumarin), purchased from Sigma Inc., St. Louis, Mo., USA can be added to a final concentration of 100 μM from a stock solution of 10 mM in dimethylsulfoxide. Reaction volumes can be 100 μl per well. After incubation for various periods of time at 37° C., the concentration of free AMC (aminomethylcoumarin) can be determined on a Perkin Elmer HTS 7000 Plus microplate reader, excitation 370 nM and emission 465 nM. Proteasome activity can be determined under conditions in which substrate hydrolysis increased linearly with time and the change in fluorescence signal is proportional to the concentration of free AMC.
Example B Assay for Activity of α-1-Chymotrypsin
In 96-well microtiter plates bovine α-chymotrypsin, purchased from Sigma Inc., can be plated at 10 ng/mL (about 2 μM catalytic sites) in 0.5 M NaCl 50 mM Hepes buffer. A fluorimetric substrate Suc-AAPF-AMC (succinyl-Ala-Ala-Pro-Phe-7-amido-4-methylcoumarin), purchased from Sigma Inc., St. Louis, Mo., USA can be added to a final concentration of 25 μM from a stock solution of 10 mM in dimethylsulfoxide. Reaction volumes are 100 μl per well. After incubation for various periods of time at room temperature, the concentration of free AMC can be determined on a Perkin Elmer HTS 7000 Plus microplate reader, excitation 370 nM and emission 465 nM. α-Chymotrypsin activity can be determined under conditions in which substrate hydrolysis increased linearly with time and the change in fluorescence signal was proportional to the concentration of free AMC.
Example C Determination of Ic50 Values for HEP and α-Chymotrypsin Inhibitors
IC50 values are typically defined as the concentration of a compound necessary to produce 50% inhibition of the enzyme's activity. IC50 values are useful indicators of the activity of a compound for its designated use. The proteasome inhibitors of the invention can be considered active if they have IC50 values of less than about 1 micromolar for inhibition of human erythrocyte proteasome (HEP). In some embodiments, the inhibitors show some specificity for HEP and the ratio of the IC50 for inhibition of bovine α-chymotrypsin versus the IC50 for inhibition of HEP, i.e, IC50 (α-Chymotripsin)/IC50 (HEP), is greater then about 100.
Inhibition of the chymotrypsin-like activity of HEP and of bovine α-chymotrypsin can be determined by incubating the enzyme with various concentrations of putative inhibitors for 15 minutes at 37° C. (or room temperature for α-chymotrypsin) prior to the addition of substrate. Each experimental condition can be evaluated in triplicate.
Compounds of the present invention are considered active in the above identified assay if their IC50 values for inhibition of HEP are less than 1000 nanoMolar. Preferably compounds of the present invention will have IC50 values for inhibition of HEP less than 100 nanoMolar. More preferably compounds of the present invention will have IC50 values for inhibition of HEP less than 10 nanoMolar.
Example D Cellular Assay for Chymotrypsin-like Activity of Proteasome in Molt-4 Cell Line
The chymotrypsin-like activity of proteasome in Molt-4 cells (human leukemia) can be assayed according to the following procedure. A brief description of the method was published previously (Harding et al., J. Immunol., 1995, 155, 1767).
Molt-4 cells are washed and resuspended in HEPES-buffered Saline (5.4 mM KCl, 120 mM NaCl, 25 mM Glucose, 1.5 mM MgSO4, 1 mM Na pyruvate, 20 mM Hepes) and plated in 96-well microtiter white plates to a final concentration of 6×106 cells/mL. Then various 5× proteasome inhibitor concentrations (or diluted DMSO for controls), prepared from 250×DMSO solutions by diluting 50-fold using HEPES-buffered saline, are added to the plate to a final 1× concentration. After 15 minutes incubation at 37° C., a fluorimetric cell permeable substrate (MeOSuc-FLF-AFC) (methoxysuccinyl-Phe-Leu-Phe-7-amido-4-trifluoromethylcoumarin) purchased from Enzyme Systems Products, catalogue number AFC-88, is added to each well at a final concentration of 25 μM from a stock solution of 20 mM in DMSO. Reaction volumes can be 100 μl per well.
The concentration of free AFC is monitored every 1.5 min for 30 min (22 cycles) on a Polastar Optima, BMG Labtechnologies microplate reader, using an excitation wavelength of 390 nm and emission wavelength of 520 nm. Proteasome activity can be determined under conditions in which substrate hydrolysis increased linearly with time and the change in fluorescent signal is proportional to the concentration of free AFC.
Example E Determination of EC50 Values for Proteasome Inhibitors in MOLT-4 Cell Line
EC50 values are typically defined as the concentration of a compound required to produce an inhibition of the enzyme's activity halfway between the minimum and the maximum response (0% and 85-90% respectively for this assay). EC50 values are useful indicators of the activity of a compound for its designated use. The compounds of the invention can be considered active if they have an EC50 of less than about 10 micromolar.
Inhibition of chymotrypsin-like activity of proteasome in Molt-4 cells is determined by incubating cells with various concentrations of putative inhibitors for 15 minutes at 37° C. prior to the addition of substrate. Each experimental condition is evaluated in triplicate.
Compounds of the present invention are considered active in the above identified assay if their EC50 values for proteasome inhibition in MOLT-4 are less than 10 microMolar. Preferably compounds of the present invention will have EC50 values for proteasome inhibition in MOLT-4 less than 2 microMolar. More preferably compounds of the present invention will have EC50 values for proteasome inhibition in MOLT-4 less than 200 nanomolar.
Example F Assay for Trypsin-like Activity of the Proteasome
The trypsin-like activity of human proteasome can be assayed as described above with the following modifications. Reactions can be carried out in Tris-glycerol buffer (pH 9.5) supplemented with 1 mM 2-mercaptoethanol, and the substrate can be a fluorogenic substrate such as benzyloxycarbonyl-Phe-Arg-AMC (100 μM).
After incubation for various periods of time at 37° C., the concentration of free AMC can be determined on a Fluoroskan II spectrofluorimeter with an excitation filter of 390 nm and an emission filter of 460 nm. Protease activity can be determined under conditions in which substrate hydrolysis increases linearly with time and the change in fluorescence is proportional to the concentration of free AMC.
Example G In vivo Inhibition of Cellular Muscle Breakdown
The effect of inhibitors on the unweighting atrophy of the soleus muscle in juvenile rats can be determined by, for example, the procedures described in Tischler, Metabolism, 1990, 39, 756. For example, juvenile female Sprague-Dawley rats (80-90 g) can be tail-cast, hind limb suspended as described in Jaspers, et al., J. Appl. Physiol., 1984, 57, 1472. The animal's hind limbs can be elevated above the floor of the cage with each animal housed individually. Animals can have free access to food and water, and can be weighed at the time of suspension and at time of termination. During the suspension period the animals can be checked daily to ensure that their toes are not touching the floor of the cage, and that there is no swelling of the tail due to the cast.
Experimental Design—Part 1
Each experiment can begin with the suspension of 20 rats which are randomly divided into 4 groups of 5 animals each. Group A can be suspended for 2 days, providing baseline data to approximate the soleus muscle size in other animals suspended for longer times. Average body weights for the groups at the outset of the study can be compared and used as a correction factor for body size differences. Group B can be a second control group which has the soleus of one limb treated with an aqueous solution of mersalyl after two days of unweighting, to demonstrate the ability to slow muscle atrophy during unweighting, for each group of animals. At 2 days after unweighting commences, an aqueous solution of mersalyl (200 nM; 4 μL/100 g initial body wt) can be injected into one soleus. The contralateral muscle can be injected with a similar volume of 0.9% saline (“Vehicle”). The animals can be maintained under Innovar-vet (10 μL/100 g body wt) tranquilization during the in situ injection procedure. After the injections, the animals can be suspended for an additional 24 hours and the soleus can be removed. Groups C and D for each experiment can be used for testing each of two different embodiments of the disclosed compounds. Animals can be treated as in group B, except that 1 mM proteasome inhibitor, contained in dimethysulfoxide (DMSO), can be injected into the soleus of one leg and DMSO only into the contralateral soleus. Thus each experiment consists of two control groups and the testing of proteasome inhibitors of the invention. The completion of five such experiments with different pairs of inhibitors provides for an “n” value of 10 for testing each inhibitor and each can be tested in two different shipments of animals.
Processing of the Soleus Muscle—Part 1
After the animal is sacrificed, the soleus can be excised, trimmed of fat and connective tissue, and carefully weighed. The muscle can then homogenized in 10% trichloroacetic acid (TCA) and the precipitated protein pelleted by centrifugation. The pellet can then be washed once with 10% TCA and once with ethanol:ether (1:1). The final pellet can be solubilized in 4 ml of 1N sodium hydroxide. The sample can be then analyzed for protein content by the biuret procedure, using albumin as a standard.
Data Analysis—Part 1
The effect of inhibitors on total muscle protein content can be examined primarily by paired comparison with the untreated contralateral muscle. The ratio of contents can be calculated and then analyzed statistically by analysis of variance (“ANOVA”). The left leg can always be the treated leg so that the protein content ratios can be compared to the non-treated control animals as well. In this way, a significant difference can be shown by comparing the protein content of the two legs, as well as the relative effectiveness of the tested inhibitors. A paired student test can also be performed for the effect of each separate treatment. The non-treated control data also provide an estimate of protein content of day 2. This allows approximation of the protein changes over the 24 hours of treatment for each of the Groups B, C, and D.
Experimental Design—Part 2
Each experiment can consist of 10 animals with groups of 5 animals being tested with one of the inhibitors for its effect on protein synthesis. Control animals are not needed for this aspect of the study as the contralateral DMSO-treated muscle serves as the paired control for the inhibitor-treated muscle. Each group can be injected as described for groups C and D in part 1. Twenty-four hours after the in situ treatment the fractional rate of protein synthesis can be analyzed in both soleus muscles. Each muscle can be injected with a 0.9% saline solution (3.5 μl/100 g final body wt) containing 3H-phenylalanine (50 mM; 1 μCi/ml). Fifteen minutes later the middle two-thirds of the muscle can be excised and the muscle can be processed as described below.
Processing of the Soleus Muscle—Part 2
The muscle can be first washed for 10 minutes in 0.84% saline containing 0.5 mM cycloheximide, to terminate protein synthesis, and 20 mM cycloleucine, to trap phenylalanine in the cell. The muscle can then be homogenized in 2.5 mL of ice-cold 2% perchloric acid. The precipitated protein can be pelleted by centrifugation. One aliquot of the supernatant can be taken for liquid scintillation counting and another aliquot can be processed for conversion of phenylalanine to phenethylamine to determine the soluble phenylalanine concentration fluorometrically. See, e.g., Garlick, et al., Biochem. J., 1980, 192, 719. These values can provide the intracellular specific activity. The specific activity of phenylalanine in the muscle protein can be determined after hydrolyzing the protein by heating in 6N HCl. The amino acids released can be solubilized in buffer. One aliquot can be taken for scintillation counting and another for analysis of phenylalanine as for the supernatant fraction. The fractional rate of protein synthesis can be calculated as: protein specific activity/intracellular specific activity x time.
Data Analysis—Part 2
Analyses of protein synthesis can be on a paired basis for each inhibitor. Student paired t test comparisons of the contralateral muscles can determine whether there is any effect of the inhibitor on protein synthesis. Protein breakdown can be calculated approximately as the fractional rate of protein synthesis (from part 2) plus the fractional rate of protein accretion (from part 1), where protein loss yields a negative value for protein accretion.
Qualitatively the ability of inhibitors to slow protein loss without affecting protein synthesis indicates a slowing of protein degradation.
Example H In vivo Investigation of Anti-Tumor Activity
Materials
The proteasome inhibitors used for in vivo studies can be formulated in an appropriate medium for intravenous (iv) or oral (po) administration. For example, for the iv administration the compounds can be administered dissolved in 0.9% NaCl, or in mixtures of 0.9% NaCl, solutol HS15 and dimethylsulfoxide, for example in the ratio 87:10:3 (v:v:v), respectively.
Cell Lines
The following human and murine tumor cell lines of different histological origin can be used to test the antitumor activity of the compounds of the invention: H460 (human, lung), A2780 (human, ovary), PC-3 (human, prostate), LoVo (human, colon), HCT116 (human, colon), BXPC3 (human, pancreatic), PANC-1 (human, pancreatic), MX-1 (human, mammary), MOLT (human, leukemia), multiple myeloma (human, myeloma), YC8 (murine, lymphoma), L1210 (murine, leukemia), 3LL (murine, lung).
Animal Species
5-6 Weeks immunocompetent or immunodeprived mice are purchased from commercial sources, for example from Harlan (Correzzana, Mi Italy). CD1 nu/nu mice are maintained under sterile conditions; sterilized cages, bedding, food and acidified water are used.
Tumor Cell Implantation and Growth
Solid tumor models of different hystotype (lung, ovary, breast, prostate, pancreatic, colon) can be transplanted subcutaneously (sc.) into the axillary region of immunocompetent mice (murine models) or in immunodeprived mice (human models). Human tumor cell lines, originally obtained from ATCC, can be adapted to grow “in vivo” as solid tumor from “in vitro culture”.
Hematological human or murine tumor models can be transplanted into different sites (iv, ip, ic or sc) in immunocompetent mice (murine tumors) or in immunodeprived mice (human leukemia, lymphoma and myeloma models), according to their highest tumor take.
Drug Treatment
Mice bearing solid (staged) or hematological tumors are randomized in experimental groups (10 mice/group). For solid tumors, an average tumor weight of 80-100 mg for each group is considered to start the treatment; mice with the smallest and largest tumors are discarded.
Experimental groups are randomly assigned to the drug treatment and to the control group. Animals can be treated iv or orally, depending on the oral bioavailability with the compounds following different treatment schedules: iv weekly or twice weekly, or by daily oral administration.
On solid tumor models, drug treatment can begin when the tumor size ranges between 80-100 mg after tumor transplantation (Day 0).
The compounds can be administered in a volume of 10 mL/Kg body weight/mouse in the appropriate solvent.
Parameters of Antitumor Activity
The following parameters can be assessed for the evaluation of the antitumor activity:
    • growth of primary solid tumor; in each mouse is monitored by caliper measurement twice weekly;
    • survival time of treated mice as compared to control mice
    • twice weekly body weight evaluation of individual mice.
The tumor growth inhibition, TWI % (percentage of primary tumor growth inhibition in comparison with vehicle treated control groups) or the Relative tumor growth inhibition, RTWI % in case of staged tumors, is evaluated one week after the last drug treatment and the Tumor weight (TW) can be calculated as follows:
TW=½ab2
where a and b are long and short diameters of the tumor mass in mm.
The antitumor activity can be determined as tumor weight inhibition (TWI %), which is calculated according to the formula:
TWI % = 100 - mean TW treated mean TW controls × 100
The RTWI % (relative percentage of primary tumor growth inhibition in comparison with vehicle treated control groups) is evaluated one week after the last drug treatment, according to the following formula:
RTWI % = 100 - mean RV of treated mice mean RV of controls mice × 100 where RV = Vt ( tumor weight on day t ) Vo ( initial tumor weight at the outset of treatment )
The Percent of Tumor Regression can be calculated as regressions in terms of relative tumor weight, determined as tumor weight at given day divided by initial tumor weight at the outset the experiment.
On haematological tumour models the antitumor activity can be determined as percentage increase of the median survival time of mice expressed as the ratio (T/C %) of the median survival time of the treated group (T) to that of the control group (C). Animals which are tumour-free at the end of the experiment (60 days after transplantation) are excluded from the calculation and considered as long term survivors (LTS).
Evaluation of Toxicity in Tumor Bearing Mice
Toxicity can be evaluated daily on the basis of the gross autopsy findings and the weight loss. Mice are considered to have died of toxicity when death occurs before the death of vehicle treated control animals, or when significant body weight loss (>20%), and/or spleen and liver size reduction are observed.
The BWC % (Body weight change %) is assessed as follow: 100−(mean body weight of mice at given day/mean body weight at start of treatment)×100. This value is determined one week after the last treatment with the test compound.
Example K In vitro Viability of Cells
The IC50 values measuring in vitro viability of cells in the presence of test compounds can be determined according to the following procedure. Cells can be seeded in 96-well plates at varying densities and then assayed using the Calcein-AM viability assay after 24 hours to determine the optimal final density for each cell type. Cells can then be seeded in 96-well plates at the determined density in 100 μL of an appropriate cell media known to one skilled in the art.
Serial dilutions of test compounds can be made so that the concentrations are twice the desired concentration to be evaluated. When 100 μL of the dilution is then added to the cells plated in 100 μL of media, a final concentration of, for example, 0, 11.7, 46.9, 187.5, 375, and 750 nM can be obtained. Compounds can be added to the plates three to four hours after seeding the cells, then the plates can be incubated at 37° C. for the desired time point (e.g., one, two, or three days).
Calcein-AM viability assays can be conducted at the desired time points as follows. Media can be aspirated using a manifold and metal plate to leave approximately 50 μL/well. The wells can be washed three times with 200 μL DPBS, aspirating each time with the manifold to leave 50 μL/well. A 8 μM solution of Calcein-AM in DPBS can be prepared and 150 μL can be added to each well. The plates can then be incubated at 37° C. for 30 minutes. After incubation, calcein can be aspirated with the manifold and cells can be washed with 200 μL DPBS as before. After final aspiration, fluorescence can be measured using a Cytofluor 2300 fluorescence plate reader. Negative controls can contain media and no cells, and experiments can be conducted in triplicate.
Example L Kinetic Experiments in vitro
Compounds of the invention can be tested for proteasome inhibitory activity using a protocol described in Rock, et al., Cell, 1994, 78, 761. According to this procedure, dissociation constants (Ki) for the equilibrium established when proteasome and test compound interact to form a complex. The reactions can be carried out using SDS-activated 20S proteasome from rabbit muscle, and the proteasome substrate can be Suc-LLVY-AMC.
Example M Inhibition of Activation of NF-κB
Compounds of the invention can be tested for inhibiting the activity of NF-κB by carrying out the assay described in Palombella, et al., Cell, 1994, 78, 773). For example, MG63 osteocarcinoma cells can be stimulated by treatment with TNF-α for designated times. Whole cell extracts can be prepared and analyzed by electrophoretic mobility shift assays using the PRDII probe from the human IFN-β gene promoter.
Pharmaceutical Formulations and Dosage Forms
When employed as pharmaceuticals, the compounds of Formula (I) can be administered in the form of pharmaceutical compositions. These compositions can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal, and can be prepared in a manner well known in the pharmaceutical art.
This invention also includes pharmaceutical compositions which contain, as the active ingredient, one or more of the compounds of Formula (I) above in combination with one or more pharmaceutically acceptable carriers. In making the compositions of the invention, the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
In preparing a formulation, the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.
Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. The compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
The compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 100 mg, more usually about 10 to about 30 mg, of the active ingredient. The term “unit dosage forms” refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
The active compound can be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention. When referring to these preformulation compositions as homogeneous, the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 500 mg of the active ingredient of the present invention.
The tablets or pills of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
The liquid forms in which the compounds and compositions of the present invention can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
The amount of compound or composition administered to a patient will vary depending upon what is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration, and the like. In therapeutic applications, compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. An amount adequate to accomplish this is referred to as “therapeutically effective amount.” Effective doses will depend on the disease condition being treated as well as by the judgement of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
The compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the compound preparations typically will be between 3and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
The therapeutic dosage of the compounds of the present invention can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of a compound of the invention in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration. For example, the compounds of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 μg/kg to about 1 g/kg of body weight per day. In some embodiments, the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day. The dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
The present invention also includes pharmaceutical kits useful, for example, in the treatment or prevention of inflammatory diseases, which comprise one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I). Such kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art. Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference cited in the present application, including patents, published patent applications, and journal articles, is incorporated herein by reference in its entirety.

Claims (21)

1. A pharmaceutical composition comprising one or more pharmaceutically acceptable carriers and a compound of Formula (I)
Figure US08283367-20121009-C00205
or pharmaceutically acceptable salt form thereof, wherein:
Q is —B(ORB)2, boronic acid, attached via the boron atom, or a cyclic boronic ester attached via the boron atom, wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3 or —CH2NR1aR1;
Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R4;
R1 is H, C1-10 alkyl, carbocyclyl, heterocyclyl, C1-10 alkyl-C(═O)—, C2-10 alkenyl-C(═O)—, C2-10 alkynyl-C(═O)—, carbocyclyl-C(═O)—, heterocyclyl-C(═O)—, carbocyclylalkyl-C(═O)—, heterocyclylalkyl-C(═O)—, C1-10 alkyl-S(═O)2—, carbocyclyl-S(═O)2—, heterocyclyl-S(═O)2—, carbocyclylalkyl-S(═O)2—, heterocyclylalkyl-S(═O)2—, C1-C10 alkyl-NHC(═O)—, carbocyclyl-NHC(═O)—, heterocyclyl-NHC(═O)—, carbocyclylalkyl-NHC(═O)—, heterocyclylalkyl-NHC(═O)—, C1-C10 alkyl-OC(═O)—, carbocyclyl-OC(═O)—, heterocyclyl-OC(═O)—, carbocyclylalkyl-OC(═O)—, heterocyclylalkyl-OC(═O)—, C1-10alkyl-NH—C(═O)—NHS(═O)2—, carbocyclyl-NH—C(═O)—NHS(═O)2—, heterocyclyl-NH—C(═O)—NHS(═O)2—, C1-10 alkyl-S(═O)2—NH—C(═O)—, carbocyclyl-S(═O)2—NH—C(═O)—, heterocyclyl-S(═O)2—NH—C(═O)—, or an amino protecting group; wherein R1 is optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3;
R1a is H; alternatively, R1a and R1 together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocyclyl group optionally substituted with 1, 2, or 3 R3;
R2 is, independently, H or C1-6 alkyl; alternatively, two R2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group;
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C (═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
R4 is, independently, selected from C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, —OR4a, —SR4a, —CN, halo, haloalkyl, —NH2, —NH(alkyl), —N(alkyl)2, —NHC(═O)O-alkyl, —NHC(═O)alkyl, —COOH, —C(═O)O-alkyl, —C(═O)alkyl, —C(O)H, —S(═O)-alkyl, —S(═O)2-alkyl, —S(═O)-aryl, —S(═O)2-aryl, carbocyclyl optionally substituted with 1, 2 or 3 R5, and heterocyclyl optionally substituted with 1, 2 or 3 R5;
R4a is H, C1-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, carbocyclyl or heterocyclyl;
R5 is, independently, selected from C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O) NH—, alkyl-C(═O)O—, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
with the proviso that when Z is —CH(OH)CH3and Q is
Figure US08283367-20121009-C00206
then Hy is other than
Figure US08283367-20121009-C00207
2. The pharmaceutical composition of claim 1 wherein Q is boronic acid or a cyclic boronic ester wherein said cyclic boronic ester contains from 6 to 10 carbon atoms and contains at least one cycloalkyl moiety.
3. The pharmaceutical composition of claim 1 wherein Q is pinanediol boronic ester.
4. The pharmaceutical composition of claim 1 wherein Z is —CH(OH)CH3.
5. The pharmaceutical composition of claim 1 wherein Z is —CH2NHR1.
6. The pharmaceutical composition of claim 1 wherein Z is —CH2NHR1 and R1 is carbocyclyl-C(═O)— or carbocyclyl-S(═O)2—, each optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3.
7. The pharmaceutical composition of claim 1 wherein Z is —CH2NHR1 and R1 is aryl-C(═O)— or aryl-S(═O)2—, each optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, F, Cl, Br, I, C1-4 haloalkyl, carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3 and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3.
8. The pharmaceutical composition of claim 1 wherein Z is —CH2NHR1 and R1 is phenyl-C(═O)— or phenyl-S(═O)2—, each optionally substituted with C1-4 alkyl, F, Cl, Br, I, or aryl.
9. The pharmaceutical composition of claim 1 wherein Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 R4.
10. The pharmaceutical composition of claim 1 wherein Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, thiazolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, or indolyl, each optionally substituted by 1, 2 or 3 R4.
11. The pharmaceutical composition of claim 1 wherein Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, thiazolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, or indolyl, each optionally substituted by 1 or 2 C1-8 alkyl, carbocyclyl optionally substituted with 1, 2 or 3 R5, or heterocyclyl optionally substituted with 1, 2 or 3 R5.
12. The pharmaceutical composition of claim 1 wherein Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, thiazolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, or indolyl, each optionally substituted by 1 or 2 methyl, ethyl, propyl, butyl, aryl optionally substituted with 1, 2 or 3 R5, or heteroaryl optionally substituted with 1, 2 or 3 R5.
13. The pharmaceutical composition of claim 1 wherein Hy is selected from:
Figure US08283367-20121009-C00208
Figure US08283367-20121009-C00209
14. The pharmaceutical composition of claim 1 wherein:
Z is —CH(OH)CH3; and
Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R4.
15. The pharmaceutical composition of claim 1 wherein:
Z is —CH(OH)CH3; and
Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 R4.
16. The pharmaceutical composition of claim 1 wherein:
Z is —CH2NHR1;
Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R4; and
R1 is carbocyclyl-C(═O)— or carbocyclyl-S(═O)2—, each optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3.
17. The pharmaceutical composition of claim 1 wherein:
Z is —CH2NHR1;
Hy is pyridyl, N-oxopyridyl, pyrimidinyl, pyrazinyl, imidazolyl, thiazolyl, oxazolyl, pyrrolyl, pyrazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, indolyl, quinazolinyl, benzoimidazolyl, benzothiazolyl, or benzoxazolyl, each optionally substituted by 1, 2 or 3 R4; and
R1 is carbocyclyl-C(═O)— or carbocyclyl-S(═O)2—, each optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, F, Cl, Br, I, C1-4 haloalkyl, carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3, and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3.
18. The pharmaceutical composition of claim 1 wherein the compound of Formula (I) is selected from:
H.1 6-Phenyl-pyrazine-2-carboxamide, N-[(1S,2R)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] H.2.1 Pyridine-2-carboxamide, N-[(1S,2R)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] H.2.2 Pyridine-3-carboxamide, N-[(1S,2R)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] H.2.3 Quinoline-2-carboxamide, N-[(1S,2R)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5- trimethyl-4,6-methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] H.2.4 Quinoxaline-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] H.2.5 5-Phenyl-pyrazine-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] H.2.6 5-Phenyl-pyridine-3-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-hydroxypropyl] H.2.7 5-Phenyl-pyridine-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-hydroxypropyl] H.2.8 4-Phenyl-pyridine-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-hydroxypropyl] H.2.9 Isoquinoline-1-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2- hydroxypropyl] H.2.10 Isoquinoline-3-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2- hydroxypropyl] H.2.11 Quinoline-3-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2- hydroxypropyl] H.1.12 5-(Thiophene-2-yl)pyridine-3-carboxamide, N-[(1S,2R)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] H.2.13 5-Phenyl-2H-pyrazole-3-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]-2- hydroxypropyl] H.2.14 1H-Indole-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-hydroxypropyl] H.2.15 6-Phenyl-pyrimidine-4-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-hydroxypropyl] H.2.16 5-Methyl-1-phenyl-1H-pyrazole-4-carboxamide, N-[(1S,2R)- 1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] H.2.17 2-Phenyl-thiazole-4-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-hydroxypropyl] H.2.18 6-(Thiophene-2-yl)pyridine-2-carboxamide, N-[(1S,2R)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-1,3,2-benzodioxaborol-2-yl]-3- methylbutyl]amino]carbonyl]-2-hydroxypropyl] H.2.19 6-Butyl-pyridine-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-hydroxypropyl] H.2.20 Pyridine-1-oxo-2-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-hydroxypropyl] H.2.21 Pyridine-1-oxo-3-carboxamide, N-[(1S,2R)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-hydroxypropyl] J.1 2-Pyrazinecarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.1 6-phenyl-2-pyridinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.2 5-butyl-2-pyridinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.3 2-pyridinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.4 3-pyridinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.5 Quinoline-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.6 Quinoxaline-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.7 6-phenyl-2-pyrazinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.8 4-phenyl-2-pyrazinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.9 5-Phenyl-pyridine-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.10 5-Phenyl-pyridine-2-carboxamide N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]-, J.2.11 4-Phenyl-pyridine-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.12 Isoquinoline-1-carboxamide N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]-, J.2.13 Isoquinoline-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.14 Quinoline-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.15 5-(Thiophene-2-yl)-pyridine-3-carboxamide, N-[(1S)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]- amino]carbonyl]-2-[(4-methylbenzoylamide)ethyl]- J.2.16 5-Phenyl-2H-pyrazole-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.17 1H-Indole-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.18 6-Phenyl-pyrimidine-4-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.19 5-Methyl-1-phenyl-1H-pyrazole-4-carboxamide N-[(1S)- 1-[[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]- amino]carbonyl]-2-[(4-methylbenzoylamide)ethyl]- J.2.20 2-Phenyl-thiazole-4-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.21 6-(Thiophene-2-yl)pyridine-2-carboxamide, N-[(1S)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]- amino]carbonyl]-2-[(4-methylbenzoylamide)ethyl]- J.2.22 6-Butyl-pyridine-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.23 6-morpholino-3-pyridinocarbonylamino, N-[(1S)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.24 Pyridine-1-oxo-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.25 Pyridine-1-oxo-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.1 6-phenyl-2-pyridinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.2 5-butyl-2-pyridinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.3 2-pyridinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.4 3-pyridinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.5 Quinoline-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.6 Quinoxaline-2-carboxamide, N-[(1S)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.7 6-phenyl-2-pyrazinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.8 4-phenyl-2-pyrazinocarboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.9 5-Phenyl-pyridine-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.10 5-Phenyl-pyridine-2-carboxamide N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]-, J.2.11 4-Phenyl-pyridine-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.12 Isoquinoline-1-carboxamide N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]-, J.2.13 Isoquinoline-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]- amino]carbonyl]-2-[(4-methylbenzoylamide)ethyl]- J.2.14 Quinoline-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.15 5-(Thiophene-2-yl)-pyridine-3-carboxamide, N-[(1S)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6- methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]- carbonyl]-2-[(4-methylbenzoylamide)ethyl]- J.2.16 5-Phenyl-2H-pyrazole-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.17 1H-Indole-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.18 6-Phenyl-pyrimidine-4-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.19 5-Methyl-1-phenyl-1H-pyrazole-4-carboxamide N-[(1S)-1- [[[(1R)-1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl- 4,6-methano-1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]- amino]carbonyl]-2-[(4-methylbenzoylamide)ethyl]- J.2.20 2-Phenyl-thiazole-4-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.21 6-(Thiophene-2-yl)pyridine-2-carboxamide, N-[(1S)-1-[[[(1R)- 1-[(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.22 6-Butyl-pyridine-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.23 6-morpholino-3-pyridinocarbonylamino, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.24 Pyridine-1-oxo-2-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- J.2.25 Pyridine-1-oxo-3-carboxamide, N-[(1S)-1-[[[(1R)-1- [(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano- 1,3,2-benzodioxaborol-2-yl]-3-methylbutyl]amino]carbonyl]- 2-[(4-methylbenzoylamide)ethyl]- K.1 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(6-Phenyl-pyrazine-2-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.1 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(pyridine- 2-carbonyl)amino]-1-oxobutyl]amino]-3-methylbutyl] K.2.2 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(pyridine- 3-carbonyl)amino]-1-oxobutyl]amino]-3-methylbutyl] K.2.3 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(quinoline- 2-carbonyl)amino]-1-oxobutyl]amino]-3-methylbutyl] K.2.4 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(quinoxaline- 2-carbonyl)amino]-1-oxobutyl]amino]-3-methylbutyl] K.2.5 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(5-phenyl- pyrazine-2-carbonyl)amino]-1-oxobutyl]amino]- 3-methylbutyl] K.2.6 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(5-phenyl-pyridine-3-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.7 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(5-phenyl-pyridine-2-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.8 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(4-phenyl-pyridine-2-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.9 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(isoquinoline-1-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.10 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(isoquinoline-3-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.11 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(quinoline-3-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.12 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(5-(thiophene-2-yl)pyridine-3-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.13 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(5-phenyl-2H-pyrazole-3-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.14 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(1H-indole-2-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.15 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(6-phenyl-pyrimidine-4-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.16 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(5-Methyl-1-phenyl-1H-pyrazole-4- carbonyl)amino]-1-oxobutyl]amino]-3-methylbutyl] K.2.17 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(2-phenyl-thiazole-4-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.18 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(6-(thiophene-2-yl)pyridine-2- carbonyl)amino]-1-oxobutyl]amino]-3-methylbutyl] K.2.19 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(6-butyl-pyridine-2-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.20 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(pyridine-1-oxo-2-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] K.2.21 Boronic acid, [(1R)-1-[[(2S,3R)-3-hydroxy-2- [(pyridine-1-oxo-3-carbonyl)amino]-1- oxobutyl]amino]-3-methylbutyl] L.1 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(pyrazinocarbonylamino)]-1- oxopropyl]amino]-3-methylbutyl] L.2.1 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(6-phenyl-pyridine-2-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.2 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(5-butyl-pyridine-2-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.3 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(pyridine-2-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.4 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(pyridine-3-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.5 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(quinoline-2-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.6 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(quinoxaline-2-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.7 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(6-phenyl-2-pyrazinocarbonylamino)]-1-oxopropyl]amino]-3- methylbutyl L.2.8 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(4-phenyl-2-pyrazinocarbonylamino)]-1-oxopropyl]amino]-3- methylbutyl L.2.9 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(5-phenyl-pyridine-3-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.10 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(5-phenyl-pyridine-2-carbonyl)amino]-1-oxopropyl]amino]- 3-methylbutyl] L.2.11 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(4-phenyl-pyridine-2-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.12 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(isoquinoline-1-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.13 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(isoquinoline-3-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.14 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(quinoline-3-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.15 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(5-(thiophene-2-yl)pyridine-3-carbonyl)amino]-1- oxopropyl]amino]-3-methylbutyl] L.2.16 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(5-phenyl-2H-pyrazole-3-carbonyl)amino]-1- oxopropyl]amino]-3-methylbutyl] L.2.17 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(1H-indole-2-carbonyl)amino]-1-oxopropyl]amino]- 3-methylbutyl] L.2.18 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(6-phenyl-pyrimidine-4-carbonyl)amino]-1-oxopropyl]amino]- 3-methylbutyl] L.2.19 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(5-Methyl-1-phenyl-1H-pyrazole-4-carbonyl)amino]-1- oxopropyl]amino]-3-methylbutyl] L.2.20 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(2-phenyl-thiazole-4-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.21 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(6-(thiophene-2-yl)pyridine-2-carbonyl)]-1- oxopropyl]amino]-3-methylbutyl L.2.22 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(6-butyl-pyridine-2-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.23 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(6-morpholino-3-pyridinocarbonylamino)]-1- oxopropyl]amino]-3-methylbutyl L.2.24 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(pyridine-1-oxo-2-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl] L.2.25 Boronic acid, [(1R)-1-[[(2S)-3-[(4-methylbenzoyl)amino]-2- [(pyridine-1-oxo-3-carbonyl)amino]-1-oxopropyl]amino]-3- methylbutyl].
19. The pharmaceutical composition of claim 1 wherein the compound of Formula (I) is a boronic anhydride.
20. The pharmaceutical composition of claim 19 wherein said boronic anhydride is a cyclic boronic anhydride.
21. A pharmaceutical composition comprising one or more pharmaceutically acceptable carriers and a compound of Formula (I)
Figure US08283367-20121009-C00210
or pharmaceutically acceptable salt form thereof, wherein:
Q is —B(ORB)2, boronic acid attached via the boron atom, or a cyclic boronic ester attached via the boron atom wherein said cyclic boronic ester contains from 2 to 20 carbon atoms, and, optionally, a heteroatom which can be N, S, or O;
RB is, independently, H, C1-4 alkyl, cycloalkyl, cycloalkylalkyl, aryl, or aralkyl;
Z is —CH(OH)CH3 or —CH2NR1aR1;
Hy is a 5- or 6-membered heterocyclic group optionally fused with an aryl or heteroaryl group, wherein said 5- or 6-membered heterocyclic group contains at least one ring-forming N atom, and wherein said Hy is optionally substituted by 1, 2 or 3 R4;
R1 is H, C1-10 alkyl, carbocyclyl, heterocyclyl, C1-10 alkyl-C(═O)—, C2-10 alkenyl-C(═O)—, C2-10 alkynyl-C(═O)—, carbocyclyl-C(═O)—, heterocyclyl-C(═O)—, carbocyclylalkyl-C(═O)—, heterocyclylalkyl-C(═O)—, C1-10 alkyl-S(═O)2—, carbocyclyl-S(═O)2—, heterocyclyl-S(═O)2—, carbocyclylalkyl-S(═O)2—, heterocyclylalkyl-S(═O)2—, C1-C10 alkyl-NHC(═O)—, carbocyclyl-NHC(═O)—, heterocyclyl-NHC(═O)—, carbocyclylalkyl-NHC(═O)—, heterocyclylalkyl-NHC(═O)—, C1-C10 alkyl-OC(═O)—, carbocyclyl-OC(═O)—, heterocyclyl-OC(═O)—, carbocyclylalkyl-OC(═O)—, heterocyclylalkyl-OC(═O)—, C1-10 alkyl-NH—C(═O)—NHS(═O)2—, carbocyclyl-NH—C(═O)—NHS(═O)2—, heterocyclyl-NH—C(═O)—NHS(═O)2—, C1-10 alkyl-S(═O)2—NH—C(═O)—, carbocyclyl-S(═O)2—NH—C(═O)—, heterocyclyl-S(═O)2—NH—C(═O)—, or an amino protecting group; wherein R1 is optionally substituted with 1, 2 or 3 substituents selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, F, Cl, Br, I, C1-4 haloalkyl, —NH2, —NHR2, —N(R2)2, —N3, —NO2, —CN, —CNO, —CNS, —C(═O)OR2, —C(═O)R2, —OC(═O)R2, —N(R2)C(═O)R2, —N(R2)C(═O)OR2, —C(═O)N(R2)2, ureido, —OR2, —SR2, —S(═O)—(C1-6 alkyl), —S(═O)2—(C1-6 alkyl), —S(═O)-aryl, —S(═O)2-aryl, —S(═O)2—N(R2)2; carbocyclyl optionally substituted with 1, 2, 3, 4 or 5 R3; and heterocyclyl optionally substituted with 1, 2, 3, 4, or 5 R3;
R1a is H; alternatively, R1a and R1 together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocyclyl group optionally substituted with 1, 2, or 3 R3;
R2 is, independently, H or C1-6 alkyl; alternatively, two R2 may be combined, together with the N atom to which they are attached, to form a 5-, 6- or 7-membered heterocyclic group;
R3 is, independently, selected from C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O)NH—, alkyl-C(═O)O—, (alkyl-O)r-alkyl, HO-(alkyl-O)r-alkyl-, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
R4 is, independently, selected from C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, —OR4a, —SR4a, —CN, halo, haloalkyl, —NH2, —NH(alkyl), —N(alkyl)2, —NHC(═O)O-alkyl, —NHC(═O)alkyl, —COOH, —C(═O)O-alkyl, —C(═O)alkyl, —C(O)H, —S(═O)-alkyl, —S(═O)2-alkyl, —S(═O)-aryl, —S(═O)2-aryl, carbocyclyl optionally substituted with 1, 2 or 3 R5, and heterocyclyl optionally substituted with 1, 2 or 3 R5;
R4a is H, C1-C20 alkyl, C2-C20 alkenyl, C2-C20 alkynyl, carbocyclyl or heterocyclyl;
R5 is, independently, selected from C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, phenyl, halo, haloalkyl, alkoxy, thialkoxy, amino, alkylamino, dialkylamino, carboxyl, alkyl-OC(═O)—, alkyl-C(═O)—, aryl-OC(═O)—, alkyl-OC(═O)NH—, aryl-OC(═O)NH—, alkyl-C(═O) NH—, alkyl-C(═O)O—, (alkyl-O)r-alkyl, HO-(alkyl-O)r-alkyl-, —OH, —SH, —CN, —N3, —CNO, —CNS, alkyl-S(═O)—, alkyl-S(═O)2—, H2NS(═O)—, and H2NS(═O)2—;
r is 0, 1, 2, 3 or 4;
with the proviso that when Z is —CH(OH)CH3 and Q is
Figure US08283367-20121009-C00211
then Hy is other than
Figure US08283367-20121009-C00212
US12/265,998 2005-02-11 2008-11-06 Proteasome inhibitors and methods of using the same Expired - Fee Related US8283367B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/265,998 US8283367B2 (en) 2005-02-11 2008-11-06 Proteasome inhibitors and methods of using the same

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US65237005P 2005-02-11 2005-02-11
US11/351,193 US7468383B2 (en) 2005-02-11 2006-02-09 Proteasome inhibitors and methods of using the same
US12/265,998 US8283367B2 (en) 2005-02-11 2008-11-06 Proteasome inhibitors and methods of using the same

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/351,193 Continuation US7468383B2 (en) 2005-02-11 2006-02-09 Proteasome inhibitors and methods of using the same

Publications (2)

Publication Number Publication Date
US20090075936A1 US20090075936A1 (en) 2009-03-19
US8283367B2 true US8283367B2 (en) 2012-10-09

Family

ID=36579482

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/351,193 Expired - Fee Related US7468383B2 (en) 2005-02-11 2006-02-09 Proteasome inhibitors and methods of using the same
US12/265,998 Expired - Fee Related US8283367B2 (en) 2005-02-11 2008-11-06 Proteasome inhibitors and methods of using the same

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/351,193 Expired - Fee Related US7468383B2 (en) 2005-02-11 2006-02-09 Proteasome inhibitors and methods of using the same

Country Status (20)

Country Link
US (2) US7468383B2 (en)
EP (1) EP1846424B1 (en)
JP (1) JP2008530108A (en)
AT (1) ATE456571T1 (en)
AU (1) AU2006213814B2 (en)
CA (1) CA2597273C (en)
CY (1) CY1109985T1 (en)
DE (1) DE602006012013D1 (en)
DK (1) DK1846424T3 (en)
ES (1) ES2339698T3 (en)
HK (1) HK1106534A1 (en)
IL (1) IL184678A (en)
MX (1) MX2007009664A (en)
MY (1) MY147921A (en)
NZ (1) NZ560607A (en)
PL (1) PL1846424T3 (en)
PT (1) PT1846424E (en)
SI (1) SI1846424T1 (en)
TW (1) TWI367886B (en)
WO (1) WO2006086600A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8541590B2 (en) 2009-12-22 2013-09-24 Cephalon, Inc. Proteasome inhibitors and processes for their preparation, purification and use

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007011760A2 (en) * 2005-07-15 2007-01-25 Kalypsys, Inc. Inhibitors of mitotic kinesin
NZ568068A (en) 2005-11-09 2011-10-28 Proteolix Inc Compounds for enzyme inhibition
NZ573759A (en) 2006-06-19 2012-03-30 Proteolix Inc Peptide epoxyketones for proteasome inhibition
EP2044055A4 (en) * 2006-07-21 2011-03-23 Takeda Pharmaceutical Amide compounds
CN105837608B (en) * 2007-08-06 2018-06-08 米伦纽姆医药公司 Proteasome inhibitor and combinations thereof and purposes
WO2009026579A1 (en) * 2007-08-23 2009-02-26 Cornell Research Foundation, Inc. Proteasome inhibitors and their use in treating pathogen infection and cancer
ES2578905T5 (en) 2007-10-04 2020-03-18 Onyx Therapeutics Inc Crystalline Peptide Epoxy Ketone Protease Inhibitors and Keto-Epoxy Amino Acid Synthesis
JP5600595B2 (en) * 2007-10-16 2014-10-01 ミレニアム ファーマシューティカルズ, インコーポレイテッド Proteasome inhibitor
EP2088205A1 (en) 2008-02-11 2009-08-12 Institut National De La Sante Et De La Recherche Medicale (Inserm) PSMB10: A diagnosis marker and therapeutic target of chronic rejection.
UA115131C2 (en) 2008-06-17 2017-09-25 Мілленніум Фармасьютікалз, Інк. BORONATE Ether COMPOUNDS AND ITS PHARMACEUTICAL RECIPES
CA2732806A1 (en) * 2008-08-04 2010-02-11 John Wityak Certain kynurenine-3-monooxygenase inhibitors, pharmaceutical compositions, and methods of use thereof
AR075090A1 (en) 2008-09-29 2011-03-09 Millennium Pharm Inc ACID DERIVATIVES 1-AMINO-2-CYCLLOBUTILETILBORONICO PROTEOSOMA INHIBITORS, USEFUL AS ANTI-BANKER AGENTS, AND PHARMACEUTICAL COMPOSITIONS THAT UNDERSTAND THEM.
WO2010048298A1 (en) 2008-10-21 2010-04-29 Proteolix, Inc. Combination therapy with peptide epoxyketones
AR075899A1 (en) 2009-03-20 2011-05-04 Onyx Therapeutics Inc PROTEASA INHIBITING CRYSTALLINE EPOXYCETON TRIPEPTIDES
EP2238973A1 (en) 2009-04-07 2010-10-13 Cephalon France Lyophilized preparations of proteasome inhibitors
JP5919196B2 (en) 2009-11-13 2016-05-18 オニキス セラピューティクス, インク.Onyx Therapeutics, Inc. Use of peptide epoxy ketones for metastasis inhibition
US8883785B2 (en) 2010-01-25 2014-11-11 Chdi Foundation, Inc. Certain kynurenine-3-monooxygenase inhibitors, pharmaceutical compositions, and methods of use thereof
BR112012022060A2 (en) * 2010-03-01 2018-05-08 Onyx Therapeutics Inc compound for inhibition of immunoproteasome
US9126997B1 (en) 2010-09-07 2015-09-08 Northwestern University Synergistic effect of glucocorticoid receptor agonists in combination with proteosome inhibitors for treating leukemia and myeloma
EP2627636B1 (en) * 2010-10-14 2015-09-02 Synthon BV Process for making bortezomib and the intermediates for the process
PT2750677T (en) 2011-08-30 2017-07-03 Chdi Foundation Inc Kynurenine-3-monooxygenase inhibitors, pharmaceutical compositions, and methods of use thereof
CA2844521A1 (en) 2011-08-30 2013-03-07 Chdi Foundation, Inc. Kynurenine-3-monooxygenase inhibitors, pharmaceutical compositions, and methods of use thereof
TW201414751A (en) 2012-07-09 2014-04-16 歐尼克斯治療公司 Prodrugs of peptide epoxy ketone protease inhibitors
WO2014072985A1 (en) * 2012-11-06 2014-05-15 Natco Pharma Limited Novel boronic acid derivatives as anti cancer agents
US9493439B1 (en) 2014-04-07 2016-11-15 University Of Kentucky Research Foundation Proteasome inhibitors
KR102322543B1 (en) * 2014-04-08 2021-11-04 앱리바 에이비 Novel cell-permeable succinate compounds
EA201692332A1 (en) 2014-05-20 2017-04-28 Милленниум Фармасьютикалз, Инк. Bordeaux Proteas Inhibitors for Use After Primary Anti-Cancer Therapy
AU2015289492B2 (en) 2014-07-17 2020-02-20 Chdi Foundation, Inc. Methods and compositions for treating HIV-related disorders
MX2019010108A (en) * 2017-02-28 2020-01-13 Chia Tai Tianqing Pharmaceutical Group Co Ltd Azetidine derivative.
GB201808149D0 (en) 2018-05-18 2018-07-11 Univ Court Univ Of Glasgow Protected amino acids
WO2020025037A1 (en) * 2018-08-02 2020-02-06 正大天晴药业集团股份有限公司 Borate of azetidine derivative

Citations (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0116678A1 (en) 1983-01-18 1984-08-29 Matth. Hohner AG Electronic musical instrument
US4499082A (en) 1983-12-05 1985-02-12 E. I. Du Pont De Nemours And Company α-Aminoboronic acid peptides
US4537773A (en) 1983-12-05 1985-08-27 E. I. Du Pont De Nemours And Company α-Aminoboronic acid derivatives
EP0293881A2 (en) 1987-06-05 1988-12-07 The Du Pont Merck Pharmaceutical Company Peptide boronic acid inhibitors of trypsin-like proteases
EP0315574A2 (en) 1987-11-05 1989-05-10 Hoechst Aktiengesellschaft Renin inhibitors
WO1989009225A1 (en) 1988-03-28 1989-10-05 The Regents Of The University Of California Nerve growth factor peptides
EP0354522A1 (en) 1988-08-12 1990-02-14 Hoechst Aktiengesellschaft Use of alpha-aminoboronic acid derivatives in prevention and treatement of viral diseases
US4963655A (en) 1988-05-27 1990-10-16 Mayo Foundation For Medical Education And Research Boron analogs of amino acid/peptide protease inhibitors
US5023236A (en) 1988-04-07 1991-06-11 Corvas, Inc. Factor VII/VIIA active site inhibitors
WO1991013904A1 (en) 1990-03-05 1991-09-19 Cephalon, Inc. Chymotrypsin-like proteases and their inhibitors
US5106948A (en) 1988-05-27 1992-04-21 Mao Foundation For Medical Education And Research Cytotoxic boronic acid peptide analogs
US5159060A (en) 1988-05-27 1992-10-27 Mayo Foundation For Medical Education And Research Cytotoxic boronic acid peptide analogs
US5242904A (en) 1987-06-05 1993-09-07 The Dupont Merck Pharmaceutical Company Peptide boronic acid inhibitors of trypsin-like proteases
US5250720A (en) 1987-06-05 1993-10-05 The Dupont Merck Pharmaceutical Company Intermediates for preparing peptide boronic acid inhibitors of trypsin-like proteases
WO1994004653A1 (en) 1992-08-14 1994-03-03 The Procter & Gamble Company Liquid detergents containing an alpha-amino boronic acid
WO1994017816A1 (en) 1993-02-10 1994-08-18 The President And Fellows Of Harvard College Role of atp-ubiquitin-dependent proteolysis in mhc-1 restricted antigen presentation and inhibitors thereof
WO1994025051A1 (en) 1993-04-30 1994-11-10 Merck & Co., Inc. Thrombin inhibitors
WO1994025049A1 (en) 1993-04-27 1994-11-10 The Du Pont Merck Pharmaceutical Company Amidino and guanidino substituted boronic acid inhibitors of trypsin-like enzymes
EP0632026A1 (en) 1993-06-30 1995-01-04 Adir Et Compagnie Alpha amino acid derivatives, a method for their preparation and pharmaceutical preparations containing them
WO1995009838A1 (en) 1993-10-01 1995-04-13 Merrell Pharmaceuticals Inc. INHIBITORS OF β-AMYLOID PROTEIN PRODUCTION
WO1995009634A1 (en) 1993-10-07 1995-04-13 The Du Pont Merck Pharmaceutical Company Electrophilic peptide analogs as inhibitors of trypsin-like enzymes
WO1995020603A1 (en) 1994-01-26 1995-08-03 Sandoz Ltd. Inhibitors of serine proteases, bearing a chelating group
WO1995025533A1 (en) 1994-03-18 1995-09-28 The President And Fellows Of Harvard College PROTEASOME REGULATION OF NF-λB ACTIVITY
WO1996011697A1 (en) 1994-10-12 1996-04-25 Merck & Co., Inc. Thrombin inhibitors
WO1996012499A1 (en) 1994-10-25 1996-05-02 The Du Pont Merck Pharmaceutical Company Amidino and guanidino substituted inhibitors of trypsin-like enzymes
WO1996013266A1 (en) 1994-10-28 1996-05-09 Proscript, Inc. Boronic ester and acid compounds, synthesis and uses
WO1996014857A1 (en) 1994-11-14 1996-05-23 Cephalon, Inc. Multicatalytic protease inhibitors
US5550262A (en) 1994-11-14 1996-08-27 Cephalon, Inc. Multicatalytic protease inhibitors
US5693617A (en) 1994-03-15 1997-12-02 Proscript, Inc. Inhibitors of the 26s proteolytic complex and the 20s proteasome contained therein
WO1998017679A1 (en) 1996-10-18 1998-04-30 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases, particularly hepatitis c virus ns3 protease
WO1998022496A2 (en) 1996-11-18 1998-05-28 F. Hoffmann-La Roche Ag Antiviral peptide derivatives
US5834487A (en) 1996-09-24 1998-11-10 Cv Therapeutics Inhibition of 26S and 20S proteasome by indanones
WO1999030707A1 (en) 1997-12-16 1999-06-24 Cephalon Inc. Multicatalytic protease inhibitors for use as anti-tumor agents
DE19914474A1 (en) 1998-03-30 1999-10-07 Hoffmann La Roche New peptide aldehyde and boronic acid derivatives are proteinase inhibitors useful for treatment of viral infections, especially hepatitis
WO2000002548A2 (en) 1998-07-10 2000-01-20 Osteoscreen Inhibitors of proteasomal activity for stimulating bone and hair growth
EP0995757A2 (en) 1998-08-26 2000-04-26 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Bivalent inhibitors of the proteasome
WO2000023614A1 (en) 1998-10-20 2000-04-27 Millennium Pharmaceuticals, Inc. Method for monitoring proteasome inhibitor drug action
US6075150A (en) 1998-01-26 2000-06-13 Cv Therapeutics, Inc. α-ketoamide inhibitors of 20S proteasome
US6096778A (en) 1997-10-07 2000-08-01 Cephalon, Inc. α-ketoamide multicatalytic protease inhibitors
WO2000064467A1 (en) 1999-04-22 2000-11-02 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Inhibition of proteasome function induces programmed cell death in endothelial cells
WO2000064863A1 (en) 1999-04-27 2000-11-02 Novartis Ag Use of 2,4-diamino-3-hydroxycarboxylic acid derivatives as proteasome inhibitors
WO2000066557A1 (en) 1999-05-03 2000-11-09 Astrazeneca Ab New compounds
WO2001002424A2 (en) 1999-07-07 2001-01-11 Du Pont Pharmaceuticals Company Peptide boronic acid inhibitors of hepatitis c virus protease
WO2001020995A1 (en) 1999-09-23 2001-03-29 Washington University Compounds directed against pilus biogenesis and activity in pathogenic bacteria; methods and compositions for synthesis thereof
WO2001028579A2 (en) 1999-10-20 2001-04-26 Osteoscreen, Inc. Inhibitors of proteasomal activity for stimulating bone and hair growth
WO2002008187A1 (en) 2000-07-21 2002-01-31 Schering Corporation Novel peptides as ns3-serine protease inhibitors of hepatitis c virus
WO2002030455A2 (en) 2000-10-12 2002-04-18 Viromics Gmbh Agents for the treatment of viral infections
WO2002059130A1 (en) 2001-01-25 2002-08-01 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Formulation of boronic acid compounds
WO2002082091A2 (en) 2001-04-09 2002-10-17 Allan Christian Shaw Method for identification of proteins from intracellular bacteria
WO2002094857A1 (en) 2001-05-23 2002-11-28 The Curators Of The University Of Missouri Inverse solid phase synthesis of peptides
WO2002096933A1 (en) 2001-05-30 2002-12-05 Novartis Ag 2-{[n-(2-amino-3-(heteroaryl or aryl)propionyl)-aminoacyl]-amino}-alkylboronic acid derivatives
US20030008828A1 (en) 1999-07-26 2003-01-09 Priestley E. Scott Novel lactam inhibitors of hepatitis C virus NS3 protease
WO2003015706A2 (en) 2001-08-16 2003-02-27 Washington State University Research Foundation Borinic acid protease inhibitors
WO2003033507A1 (en) 2001-10-12 2003-04-24 Kyorin Pharmaceutical Co., Ltd. Benzylmalonic acid derivatives and proteasome inhibitors contaiing the same
WO2003059898A2 (en) 2002-01-08 2003-07-24 Eisai Co. Ltd. Eponemycin and epoxomicin analogs and uses thereof
WO2005016659A1 (en) 2003-08-13 2005-02-24 Seiko Precision Inc. Method and device for manufacturing card
WO2005021558A2 (en) 2003-08-14 2005-03-10 Cephalon, Inc. Proteasome inhibitors and methods of using the same
WO2009020448A1 (en) 2007-08-06 2009-02-12 Millennium Pharmaceuticals, Inc. Proteasome inhibitors

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100399359B1 (en) * 2001-07-07 2003-09-26 삼성전자주식회사 Charge pump circuit

Patent Citations (84)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0116678A1 (en) 1983-01-18 1984-08-29 Matth. Hohner AG Electronic musical instrument
US4499082A (en) 1983-12-05 1985-02-12 E. I. Du Pont De Nemours And Company α-Aminoboronic acid peptides
US4537773A (en) 1983-12-05 1985-08-27 E. I. Du Pont De Nemours And Company α-Aminoboronic acid derivatives
EP0293881A2 (en) 1987-06-05 1988-12-07 The Du Pont Merck Pharmaceutical Company Peptide boronic acid inhibitors of trypsin-like proteases
US5242904A (en) 1987-06-05 1993-09-07 The Dupont Merck Pharmaceutical Company Peptide boronic acid inhibitors of trypsin-like proteases
US5250720A (en) 1987-06-05 1993-10-05 The Dupont Merck Pharmaceutical Company Intermediates for preparing peptide boronic acid inhibitors of trypsin-like proteases
US5187157A (en) 1987-06-05 1993-02-16 Du Pont Merck Pharmaceutical Company Peptide boronic acid inhibitors of trypsin-like proteases
EP0315574A2 (en) 1987-11-05 1989-05-10 Hoechst Aktiengesellschaft Renin inhibitors
US5169841A (en) 1987-11-05 1992-12-08 Hoechst Aktiengesellschaft Renin inhibitors
WO1989009225A1 (en) 1988-03-28 1989-10-05 The Regents Of The University Of California Nerve growth factor peptides
US5023236A (en) 1988-04-07 1991-06-11 Corvas, Inc. Factor VII/VIIA active site inhibitors
US4963655A (en) 1988-05-27 1990-10-16 Mayo Foundation For Medical Education And Research Boron analogs of amino acid/peptide protease inhibitors
US5106948A (en) 1988-05-27 1992-04-21 Mao Foundation For Medical Education And Research Cytotoxic boronic acid peptide analogs
US5159060A (en) 1988-05-27 1992-10-27 Mayo Foundation For Medical Education And Research Cytotoxic boronic acid peptide analogs
EP0354522A1 (en) 1988-08-12 1990-02-14 Hoechst Aktiengesellschaft Use of alpha-aminoboronic acid derivatives in prevention and treatement of viral diseases
WO1991013904A1 (en) 1990-03-05 1991-09-19 Cephalon, Inc. Chymotrypsin-like proteases and their inhibitors
WO1994004653A1 (en) 1992-08-14 1994-03-03 The Procter & Gamble Company Liquid detergents containing an alpha-amino boronic acid
US5580486A (en) 1992-08-14 1996-12-03 The Procter & Gamble Company Liquid detergents containing an α-amino boronic acid
WO1994017816A1 (en) 1993-02-10 1994-08-18 The President And Fellows Of Harvard College Role of atp-ubiquitin-dependent proteolysis in mhc-1 restricted antigen presentation and inhibitors thereof
WO1994025049A1 (en) 1993-04-27 1994-11-10 The Du Pont Merck Pharmaceutical Company Amidino and guanidino substituted boronic acid inhibitors of trypsin-like enzymes
US5658885A (en) 1993-04-27 1997-08-19 The Dupont Merck Pharmaceutical Company Amidino and guanidino substituted boronic acid inhibitors of trypsin-like enzymes
WO1994025051A1 (en) 1993-04-30 1994-11-10 Merck & Co., Inc. Thrombin inhibitors
EP0632026A1 (en) 1993-06-30 1995-01-04 Adir Et Compagnie Alpha amino acid derivatives, a method for their preparation and pharmaceutical preparations containing them
US5470864A (en) 1993-06-30 1995-11-28 Adir Et Compagnie α-amino acid compounds
US5585390A (en) 1993-06-30 1996-12-17 Adir Et Compagnie Alpha-amino acid compounds
WO1995009838A1 (en) 1993-10-01 1995-04-13 Merrell Pharmaceuticals Inc. INHIBITORS OF β-AMYLOID PROTEIN PRODUCTION
WO1995009634A1 (en) 1993-10-07 1995-04-13 The Du Pont Merck Pharmaceutical Company Electrophilic peptide analogs as inhibitors of trypsin-like enzymes
WO1995020603A1 (en) 1994-01-26 1995-08-03 Sandoz Ltd. Inhibitors of serine proteases, bearing a chelating group
US5693617A (en) 1994-03-15 1997-12-02 Proscript, Inc. Inhibitors of the 26s proteolytic complex and the 20s proteasome contained therein
WO1995025533A1 (en) 1994-03-18 1995-09-28 The President And Fellows Of Harvard College PROTEASOME REGULATION OF NF-λB ACTIVITY
WO1996011697A1 (en) 1994-10-12 1996-04-25 Merck & Co., Inc. Thrombin inhibitors
WO1996012499A1 (en) 1994-10-25 1996-05-02 The Du Pont Merck Pharmaceutical Company Amidino and guanidino substituted inhibitors of trypsin-like enzymes
US6548668B2 (en) 1994-10-28 2003-04-15 Millennium Pharmaceuticals, Inc. Boronic ester and acid compounds, synthesis and uses
US6083903A (en) 1994-10-28 2000-07-04 Leukosite, Inc. Boronic ester and acid compounds, synthesis and uses
US6297217B1 (en) 1994-10-28 2001-10-02 Millennium Pharmaceuticals, Inc. Boronic ester and acid compounds, synthesis and uses
WO1996013266A1 (en) 1994-10-28 1996-05-09 Proscript, Inc. Boronic ester and acid compounds, synthesis and uses
US6066730A (en) 1994-10-28 2000-05-23 Proscript, Inc. Boronic ester and acid compounds, synthesis and uses
US6465433B1 (en) 1994-10-28 2002-10-15 Millennium Pharmaceuticals, Inc. Boronic ester and acid compounds, synthesis and uses
US5780454A (en) 1994-10-28 1998-07-14 Proscript, Inc. Boronic ester and acid compounds
US20020173488A1 (en) 1994-10-28 2002-11-21 Millennium Pharmaceuticals, Inc. Boronic Ester and acid compounds, synthesis and uses
US5614649A (en) 1994-11-14 1997-03-25 Cephalon, Inc. Multicatalytic protease inhibitors
US5990083A (en) 1994-11-14 1999-11-23 Cephalon, Inc. Multicatalytic protease inhibitors
US5830870A (en) 1994-11-14 1998-11-03 Cephalon, Inc. Multicatalytic protease inhibitors
US5550262A (en) 1994-11-14 1996-08-27 Cephalon, Inc. Multicatalytic protease inhibitors
WO1996014857A1 (en) 1994-11-14 1996-05-23 Cephalon, Inc. Multicatalytic protease inhibitors
US5834487A (en) 1996-09-24 1998-11-10 Cv Therapeutics Inhibition of 26S and 20S proteasome by indanones
WO1998017679A1 (en) 1996-10-18 1998-04-30 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases, particularly hepatitis c virus ns3 protease
WO1998022496A2 (en) 1996-11-18 1998-05-28 F. Hoffmann-La Roche Ag Antiviral peptide derivatives
US6096778A (en) 1997-10-07 2000-08-01 Cephalon, Inc. α-ketoamide multicatalytic protease inhibitors
US6310057B1 (en) 1997-10-07 2001-10-30 Cephalon, Inc. α-ketoamide multicatalytic protease inhibitors
WO1999030707A1 (en) 1997-12-16 1999-06-24 Cephalon Inc. Multicatalytic protease inhibitors for use as anti-tumor agents
US20010012854A1 (en) 1997-12-16 2001-08-09 Robert Siman Multicatalytic protease inhibitors for use as anti-tumor agents
US6075150A (en) 1998-01-26 2000-06-13 Cv Therapeutics, Inc. α-ketoamide inhibitors of 20S proteasome
DE19914474A1 (en) 1998-03-30 1999-10-07 Hoffmann La Roche New peptide aldehyde and boronic acid derivatives are proteinase inhibitors useful for treatment of viral infections, especially hepatitis
US6372883B1 (en) 1998-03-30 2002-04-16 Hoffmann-La Roche Inc. Antiviral medicaments
WO2000002548A2 (en) 1998-07-10 2000-01-20 Osteoscreen Inhibitors of proteasomal activity for stimulating bone and hair growth
EP0995757A2 (en) 1998-08-26 2000-04-26 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Bivalent inhibitors of the proteasome
WO2000023614A1 (en) 1998-10-20 2000-04-27 Millennium Pharmaceuticals, Inc. Method for monitoring proteasome inhibitor drug action
WO2000064467A1 (en) 1999-04-22 2000-11-02 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Inhibition of proteasome function induces programmed cell death in endothelial cells
WO2000064863A1 (en) 1999-04-27 2000-11-02 Novartis Ag Use of 2,4-diamino-3-hydroxycarboxylic acid derivatives as proteasome inhibitors
WO2000066557A1 (en) 1999-05-03 2000-11-09 Astrazeneca Ab New compounds
WO2001002424A2 (en) 1999-07-07 2001-01-11 Du Pont Pharmaceuticals Company Peptide boronic acid inhibitors of hepatitis c virus protease
US20030008828A1 (en) 1999-07-26 2003-01-09 Priestley E. Scott Novel lactam inhibitors of hepatitis C virus NS3 protease
WO2001020995A1 (en) 1999-09-23 2001-03-29 Washington University Compounds directed against pilus biogenesis and activity in pathogenic bacteria; methods and compositions for synthesis thereof
WO2001028579A2 (en) 1999-10-20 2001-04-26 Osteoscreen, Inc. Inhibitors of proteasomal activity for stimulating bone and hair growth
WO2002008187A1 (en) 2000-07-21 2002-01-31 Schering Corporation Novel peptides as ns3-serine protease inhibitors of hepatitis c virus
WO2002030455A2 (en) 2000-10-12 2002-04-18 Viromics Gmbh Agents for the treatment of viral infections
US20020188100A1 (en) 2001-01-25 2002-12-12 Millennium Pharmaceuticals, Inc. Formulation of boronic acid compounds
WO2002059131A1 (en) 2001-01-25 2002-08-01 Millennium Pharmaceuticals, Inc. Formulation of boronic acid compounds
WO2002059130A1 (en) 2001-01-25 2002-08-01 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Formulation of boronic acid compounds
WO2002082091A2 (en) 2001-04-09 2002-10-17 Allan Christian Shaw Method for identification of proteins from intracellular bacteria
WO2002094857A1 (en) 2001-05-23 2002-11-28 The Curators Of The University Of Missouri Inverse solid phase synthesis of peptides
WO2002096933A1 (en) 2001-05-30 2002-12-05 Novartis Ag 2-{[n-(2-amino-3-(heteroaryl or aryl)propionyl)-aminoacyl]-amino}-alkylboronic acid derivatives
WO2003015706A2 (en) 2001-08-16 2003-02-27 Washington State University Research Foundation Borinic acid protease inhibitors
WO2003033507A1 (en) 2001-10-12 2003-04-24 Kyorin Pharmaceutical Co., Ltd. Benzylmalonic acid derivatives and proteasome inhibitors contaiing the same
US20050101781A1 (en) 2002-01-08 2005-05-12 Sergei Agoulnik Eponemycin and epoxomicin analogs and uses thereof
WO2003059898A2 (en) 2002-01-08 2003-07-24 Eisai Co. Ltd. Eponemycin and epoxomicin analogs and uses thereof
WO2005016659A1 (en) 2003-08-13 2005-02-24 Seiko Precision Inc. Method and device for manufacturing card
WO2005021558A2 (en) 2003-08-14 2005-03-10 Cephalon, Inc. Proteasome inhibitors and methods of using the same
US20050107307A1 (en) 2003-08-14 2005-05-19 Raffaella Bernadini Proteasome inhibitors and methods of using the same
US7576206B2 (en) 2003-08-14 2009-08-18 Cephalon, Inc. Proteasome inhibitors and methods of using the same
US7915236B2 (en) 2003-08-14 2011-03-29 Cephalon, Inc. Proteasome inhibitors and methods of using the same
US8058262B2 (en) 2003-08-14 2011-11-15 Cephalon, Inc. Proteasome inhibitors and methods of using the same
WO2009020448A1 (en) 2007-08-06 2009-02-12 Millennium Pharmaceuticals, Inc. Proteasome inhibitors

Non-Patent Citations (54)

* Cited by examiner, † Cited by third party
Title
"Protective Groups in Organic Synthesis", 3rd edition, edited by Greene and Wuts, pp. 531-537, published in 1999.
Adams et al., "Potent and Selective Inhibitors of The Proteasome: Dipeptidyl Boronic Acids," Bioorganic & Medicinal Chemistry Letters, 1998, vol. 8, Issue 4, pp. 333-338.
Aoyagi et al., "Structures and activities of protease inhibitors of microbial origin,"Proteases and Biological Control, 1975, pp. 429-454.
Attwood et al., "The Design and Synthesis of Potent Inhibitors of Hepatitis C Virus NS3-4A Proteinase," Antiviral Chemistry & Chemotherapy, 1999, vol. 10, Issue 5, pp. 259-273.
Cecil's Textbook of Medicine (2000). Principles of Cancer Therapy. *
Christie et al., "Alzheimer's Disease: Correlation of The Suppression of beta-Amyloid Peptide Secretion From Cultured Cells With Inhibition of The Chymotrypsin-Like Activity of the Proteasome," J. of Neurochemistry, 1999, vol. 73, Issue 1, pp. 195-204.
Christie et al., "Alzheimer's Disease: Correlation of The Suppression of β-Amyloid Peptide Secretion From Cultured Cells With Inhibition of The Chymotrypsin-Like Activity of the Proteasome," J. of Neurochemistry, 1999, vol. 73, Issue 1, pp. 195-204.
Dick et al., "Degradation of oxidized insulin B chain by the multiproteinase complex macropain (proteasome)," Biochemistry, Mar. 12, 1991;30(10):2725-34.
Dudson et al., "Solid Phase Synthesis of Aminoboronic Acids: Potent Inhibitors of the Hepatitis C Virus NS3 Proteinase," Bioorganic & Medicinal Chemistry Letters, 2000, vol. 10, No. 14, pp. 1577-1579.
European Opposition-Auxiliary Request 1 Replacement Page, Sep. 21, 2011.
European Opposition-Auxiliary Request 1, Sep. 5, 2011.
European Opposition-Auxiliary Request 2 Replacement Page, Sep. 21, 2011.
European Opposition-Auxiliary Request 2, Sep. 5, 2011.
European Opposition-Main Request Replacement Page, Sep. 21, 2011.
European Opposition-Main Request, Sep. 5, 2011.
European Opposition-Opponent's Reply, Sep. 19, 2011.
European Opposition-Proprietor's Amended Claims, Jan. 17, 2011.
European Opposition-Proprietor's Response 1 to Opponent's Reply, Sep. 21, 2011.
European Opposition-Proprietor's Response 2 to Opponent's Reply, Sep. 21, 2011.
European Opposition-Proprietor's Response to Notice of Opposition, Jan. 17, 2011.
European Opposition-Proprietor's Submissions, Sep. 5, 2011.
Evrard-Todeschi et al., "Conformations in Soluation and Bound to Bacterial Ribonsomes of Ketolides, HMR 3647 (Telithromycin) and RU 72366: A New Class of Highly Potent Antibacterials," Bioorg. Med. Chem., Jul. 2000;8(7):1579-97.
Fenteany et al., "A beta-lactone related to lactacystin induces neurite outgrowth in a neuroblastoma cell line and inhibits cell cycle progression in an osteosarcoma cell line," Proc Natl Aced Sci, Apr. 12, 1994;91(8):3358-62.
Fenteany et al., "Inhibition of proteasome activities and subunit-specific amino-terminal threonine modification by lactacystin," Science, May 5, 1995;268(5211):726-31.
Garcia et al., "A new structural class of selective and non-covalent inhibitors of the chymotrypsin-like activity of the 205 proteasome," Bioorg Med Chem Lett., May 21, 2001;11(10):1317-9.
Gardner et al., "Characterization of peptidyl boronic acid inhibitors of mammalian 20 S and 26 S proteasomes and their inhibition of proteasomes in cultured cells," Biochem J., Mar. 1, 2000;346 Pt 2:447-54.
Goldberg et al., "Proteolysis, proteasomes and antigen presentation," Nature. Jun. 4, 1992;357(6377):375-9.
Goldberg et al., "The mechanism and functions of ATP-dependent proteases in bacterial and animal cells," Eur J Biochem. Jan. 15, 1992;203(1-2):9-23.
Iqbal et al., "Potent alpha-Ketocarbonyl and Boronic Ester Derived Inhibitors of Proteasome," Bioorg. Med. Chem. Lett., 1996, vol. 6, p. 287-290.
Iqbal et al., "Potent inhibitors of proteasome," J Med Chem., Jun. 23, 1995;38(13):2276-7.
Iqbal et al., "Potent α-Ketocarbonyl and Boronic Ester Derived Inhibitors of Proteasome," Bioorg. Med. Chem. Lett., 1996, vol. 6, p. 287-290.
Kettner et al., "Inhibition of the serine proteases leukocyte elastase, pancreatic elastase, cathepsin G, and chymotrypsin by peptide boronic acids," J Biol Chem. Dec. 25, 1984;259(24)15106-14.
King et al. How proteolysis drives the cell cycle. Science, vol. 274, Dec. 6, 1996. *
Kisselev et al., "Proteasome inhibitors: from research tools to drug candidates," Chem. Biol., Aug. 2001;8(8):739-58.
Kupperman et al., "Evaluation of the Proteasome Inhibitor MLN9708 in Preclinical Models of Human Cancer," Cancer Research (2010), vol. 70(5), pp. 1970-1980.
Li et al., "Isolation and characterization of a novel endogenous inhibitor of the proteasome," Biochemistry, Oct. 8, 1991;30(40)9709-15.
Matteson et al., "99% Chirally Selective Synthesis Via Pinanediol Boronic Esters: Insect Pheromones, Diols, and an Amino Alcohol," J. Am. Chem. Soc., 1986, vol. 108, pp. 810-819.
Murakami et al., "Endogenous inhibitor of nonlysosomal high molecular weight protease and calcium-dependent protease," Proc Natl Acad Sci., Oct. 1986:83(20):7588-92.
Nabel, G.J. 2001, Challenges and opportunities for development of an AIDS vaccine. Nature, 410: 1002-1007. *
Nkemgu-Njinkeng et al., "Antitrypanosomal Activities of Proteasome Inhibitors," Antimicrob. Agents Chemother., Jun. 2002;46(6):2038-40.
Notice of Opposition against European Patent EP1660507 B9 (corresponding to instant application). May 4, 2010.
Orlowski et al., "The multicatalytic proteinase complex, a major extralysosomal proteolytic system," Biochemistry, Nov. 13, 1990;29(45):10289-97.
Pantaleo & Koup. Correlates of immune protection in HIV-1 infection: what we know, what we don't know and what we should know. Nature Medicine, vol. 10, No. 8, Aug. 2004. *
Purandare et al., "Identification of a Potent and Rapidly Reversible Inhibitor of the 20S-Proteasome," Bioorganic & Medicinal Chemistry Letters, 2004, pp. 1-4.
Rivett et al., "The multicatalytic proteinase of mammalian cells," Arch Biochem Biophys., Jan. 1989;268(1):1-8.
Rivett et al., "The multicatalytic proteinase. Multiple proteolytic activities," J Biol Chem., Jul. 25, 1989;264(21):12215-9.
Rock et al., "Inhibitors of the proteasome block the degradation of most cell proteins and the generation of peptides presented on MHC class 1 molecules," Cell. Sep. 9, 1994;78(5):761-71.
Simov, Biljana Peric, et al., Chiral Carbanions, Part 4: Borylation of (Trimethylsilyl)methyl N,N-Dialkyl-carbamates-Diastereoselectivity and Structural Studies, Synthesis 2004, No. 16, pp. 2704-2710.
Song et al., The Journal of Biological Chemistry, 1999, vol. 274, pp. 1677-1682.
Tanaka et al., "Proteasomes: protein and gene structures," New Biol., 1992, vol. 4(3):173-187.
Tsubuki et al., "Purification and characterization of a Z-Leu-Leu-Leu-MCA degrading protease expected to regulate neurite formation: a novel catalytic activity in proteasome," Biochem Biophys Res Commun., Nov. 15, 1993;196(3):1195-201.
Vinitsky et al., "Inhibition of the chymotrypsin-like activity of the pituitary multicatalytic proteinase complex," Biochemistry, Oct. 6, 1992;31(39):9421-8.
Wu et al., "Proteasome Inhibitors Stimulate Activator Protein-1 Pathway Via Reactive Oxygen Species Production," FEBS Letters, 2002, vol. 526, Issue 1, pp. 101-105.
Zembower et al., "Versatile Synthetic Ligands for Affinity Chromatography of Serine Proteinases," Int'l J. of Peptide & Protein Research, 1996, vol. 47, Issue 5, pp. 405-413.

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8541590B2 (en) 2009-12-22 2013-09-24 Cephalon, Inc. Proteasome inhibitors and processes for their preparation, purification and use

Also Published As

Publication number Publication date
DE602006012013D1 (en) 2010-03-18
EP1846424B1 (en) 2010-01-27
WO2006086600A1 (en) 2006-08-17
US7468383B2 (en) 2008-12-23
IL184678A0 (en) 2007-12-03
JP2008530108A (en) 2008-08-07
PL1846424T3 (en) 2010-06-30
CA2597273C (en) 2013-12-17
TWI367886B (en) 2012-07-11
US20090075936A1 (en) 2009-03-19
CY1109985T1 (en) 2014-09-10
PT1846424E (en) 2010-04-15
CA2597273A1 (en) 2006-08-17
ES2339698T3 (en) 2010-05-24
AU2006213814B2 (en) 2011-10-13
IL184678A (en) 2012-05-31
EP1846424A1 (en) 2007-10-24
HK1106534A1 (en) 2008-03-14
MX2007009664A (en) 2007-09-26
ATE456571T1 (en) 2010-02-15
TW200640936A (en) 2006-12-01
SI1846424T1 (en) 2010-05-31
NZ560607A (en) 2009-11-27
AU2006213814A1 (en) 2006-08-17
MY147921A (en) 2013-02-15
DK1846424T3 (en) 2010-05-17
US20060189806A1 (en) 2006-08-24

Similar Documents

Publication Publication Date Title
US8283367B2 (en) Proteasome inhibitors and methods of using the same
US9233115B2 (en) Proteasome inhibitors and methods of using the same
US7223745B2 (en) Proteasome inhibitors and methods of using the same

Legal Events

Date Code Title Description
STCF Information on status: patent grant

Free format text: PATENTED CASE

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FPAY Fee payment

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20201009